WO2006024931A2 - Therapeutic combinations comprising a selective estrogen receptor modulator and a selective androgen receptor modulator - Google Patents

Therapeutic combinations comprising a selective estrogen receptor modulator and a selective androgen receptor modulator Download PDF

Info

Publication number
WO2006024931A2
WO2006024931A2 PCT/IB2005/002592 IB2005002592W WO2006024931A2 WO 2006024931 A2 WO2006024931 A2 WO 2006024931A2 IB 2005002592 W IB2005002592 W IB 2005002592W WO 2006024931 A2 WO2006024931 A2 WO 2006024931A2
Authority
WO
WIPO (PCT)
Prior art keywords
benzonitrile
amino
trifluoromethyl
ethyl
methyl
Prior art date
Application number
PCT/IB2005/002592
Other languages
French (fr)
Other versions
WO2006024931A3 (en
Inventor
Hua Zhu Ke
David Duane Thompson
Original Assignee
Pfizer Products Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Products Inc. filed Critical Pfizer Products Inc.
Publication of WO2006024931A2 publication Critical patent/WO2006024931A2/en
Publication of WO2006024931A3 publication Critical patent/WO2006024931A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • 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

Definitions

  • the present invention relates to a pharmaceutical combination of a selective estrogen receptor modulator (SERM) and a selective androgen receptor modulator (SARM) that is useful for the prevention and treatment of musculoskeletal disorders and female sexual dysfunction.
  • SERM selective estrogen receptor modulator
  • SARM selective androgen receptor modulator
  • the present invention also relates to the use of such combinations to treat female sexual dysfunction and musculoskeletal disorders, including osteoporosis, osteoporotic fracture, low bone mass, frailty and the like in mammals, including humans.
  • this invention relates to a combination of cis-6-phenyl-5-(4-(2-pyrrolidin- 1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol, or a pharmaceutically acceptable salt thereof and a selective androgen receptor modulator.
  • Osteoporosis is a systemic skeletal disease, characterized by low bone mass and deterioration of bone tissue with a consequent increase in bone fragility and susceptibility to fracture.
  • the condition affects more than 25 million people and causes more than 1.3 million fractures each year, including 500,000 spine, 250,000 hip and 240,000 wrist fractures annually.
  • Hip fractures are the most serious, and are associated with a 20% excess mortality in the year following fracture, and over 50% of the survivors being incapacitated.
  • the elderly are at greatest risk of osteoporosis, and the problem is therefore expected to increase significantly during the next several decades with the aging of the population and by increasing longevity.
  • the cost of managing fractures is substantial as approximately $13.8 billion dollars were spent in the U.S. in 1995 alone.
  • Worldwide fracture incidence is forecast to increase three-fold over the next 60 years, and one study estimates that there will be 4.5 million hip fractures worldwide in 2050. The direct as well as indirect costs of fractures are therefore expected to increase correspondingly.
  • Estrogen is an agent useful for preventing osteoporosis or postmenopausal bone loss in women.
  • Black, et al., in EP 0605193A1 report that estrogen, particularly when taken orally, lowers plasma levels of LDL and raises those of the beneficial high density lipoproteins (HDL's).
  • Raloxifene (LY139481 HCI) Prevents Bone Loss and Reduces Serum Cholesterol Without Causing Uterine Hypertrophy in Ovariectomized Rats, J. Clin. Invest., 1994, 93, 63-69 and Delmas, P.D. et al., Effects of Raloxifene on Bone Mineral Density, Serum Cholesterol Concentration, and Uterine Endometrium in Postmenopausal Women, New England Journal of Medicine, 1997, 337, 1641-1647].
  • tamoxifen 1-(4-b-dimethylaminoethoxyphenyl)-1 ,2-diphenyl-but-1-ene
  • tamoxifen 1-(4-b-dimethylaminoethoxyphenyl)-1 ,2-diphenyl-but-1-ene
  • is an antiestrogen that is proposed as an osteoporosis agent which has a palliative effect on breast cancer, but is reported to have some estrogenic activity in the uterus.
  • U.S. Patent No. 5,254,595 discloses agents such as droloxifene, which prevent bone loss, reduce the risk of fracture and are useful for the treatment of osteoporosis.
  • Female sexual dysfunction includes hypoactive sexual desire disorder, sexual anhedonia and dyspareunia. Proper sexual functioning in women depends on the sexual response cycle, which consists of an anticipatory mental set (sexual motive state or state of desire), effective vasocongestive arousal (swelling and lubrication), orgasm, and resolution.
  • orgasm is accompanied by contractions (not always subjectively experienced as such) of the muscles of the outer third of the vagina.
  • Generalized muscular tension, perineal contractions, and involuntary pelvic thrusting (every 0.8 sec) usually occur. Orgasm is followed by resolution-a sense of general pleasure, well-being, and muscular relaxation. During this phase, women may be able to respond to additional stimulation almost immediately.
  • the sexual response cycle is mediated by a delicate, balanced interplay between the sympathetic and parasympathetic nervous systems.
  • Vasocongestion is largely mediated by parasympathetic (cholinergic) outflow; orgasm is predominantly sympathetic (adrenergic).
  • cholinergic cholinergic
  • orgasm is predominantly sympathetic (adrenergic).
  • These responses are easily inhibited by cortical influences or by impaired hormonal, neural, or vascular mechanisms.
  • Disorders of sexual response may involve one or more of the cycle's phases. Generally, both the subjective components of desire, arousal, and pleasure and the objective components of performance, vasocongestion, and orgasm are disturbed, although any may be affected independently.
  • Sexual dysfunctions may be lifelong (no effective performance ever, generally due to intrapsychic conflicts) or acquired (after a period of normal function); generalized or limited to certain situations or certain partners; and total or partial.
  • FSAD is a highly prevalent sexual disorder affecting pre-, peri- and post menopausal ( ⁇ HRT) women, it is associated with concomitant disorders such as depression, cardiovascular diseases, diabetes and UG disorders.
  • ⁇ HRT menopausal
  • the primary consequences of FSAD are lack of engorgement/swelling, lack of lubrication and lack of pleasurable genital sensation.
  • the secondary consequences of FSAD are reduced sexual desire, pain during intercourse and difficulty in achieving an orgasm. It has recently been hypothesized that there is a vascular basis for at least a proportion of patients with symptoms of FSAD
  • Estrogen increases vasodilatation and inhibits the response of blood vessels to injury and the development of atherosclerosis. Estrogen-induced vasodilatation occurs 5 to 20 minutes after estrogen has been administered and is not dependent on changes in gene expression; this action of estrogen is sometimes referred to as "nongenomic.”
  • the estrogen-induced inhibition of the response to vascular injury and the preventive effect of estrogen against atherosclerosis occur over a period of hours or days after estrogen treatment and are dependent on changes in gene expression in the vascular tissues; these actions are sometimes referred to as "genomic.”
  • Cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)- 5,6,7,8,-tetrahydronaphthalene-2-ol, anc j more particularly (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol is an orally active, highly potent SERM which prevents bone loss, decreases total serum cholesterol, and does not have estrogen-like uterine stimulating effects in OVX rats.
  • 5,948,809 discloses (-)-cis-6-phenyl-5-(4-(2- pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol, tartrate salt.
  • U.S. Patent Application No. 09/833,169, filed April 11 , 2001 discloses methods of treating female sexual dysfunction with compounds such as cis-6-phenyl-5-(4-(2-pyrrolidin-1 -yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2- ol, of the Formula immediately hereinabove.
  • a selective androgen receptor modulator is a compound that possesses androgenic activity and which exerts tissue-selective effects.
  • SARM compounds can function as androgen receptor agonists, partial agonists, partial antagonists or antagonists.
  • suitable SARMs include compounds such as cyproterone acetate, chlormadinone, flutamide, hydroxyflutamide, bicalutamide, nilutamide, spironolactone, 4-(trifluoromethyl)-2(1 H)-pyrrolidino[3,2-g] quinoline derivatives, 1 ,2-dihydropyridino [5,6- g]quinoline derivatives and piperidino[3,2-g]quinolinone derivatives.
  • Cypterone also known as (ib ⁇ bJ- ⁇ -chloro-i ⁇ -dihydro- ⁇ -hydroxy-S'l-l-cyclopropa ⁇ ⁇ pregna- 1 ,4,6-triene-3,20-dione is disclosed in U.S. Patent 3,234,093.
  • Chlormadinone also known as 17- (acetyloxy)-6-chloropregna-4,6-diene-3,20-dione, in its acetate form, acts as an anti-androgen and is disclosed in U.S. Patent 3,485,852.
  • Nilutamide also known as 5,5-dimethyl-3-[4-nito-3-(trifluoromethyl) phenyl]-2,4-imidazolidinedione and by the trade name Nilandron ® is disclosed in U.S. Patent 4,097,578.
  • Flutamide also known as 2-methyl-N-[4-nitro-3-(trifluoromethyl) phenyl] propanamide and the trade name Eulexin ® is disclosed in U.S. Patent 3,847,988.
  • Bicalutamide also known as 4'-cyano-a',a',a'-trifluoro-3-(4- fluorophenylsulfonyl)-2-hydroxy-2-methylpropiono-m-toluidide and the trade name Casodex ® is disclosed in EP-100172.
  • the enantiomers of biclutamide are discussed by Tucker and Chesterton, J. Med. Chem. 1988, 31 , 885-887.
  • Hydroxyflutamide a known androgen receptor antagonist in most tissues, has been suggested to function as a SARM for effects on IL-6 production by osteoblasts as disclosed in Hofbauer et al. J. Bone Miner. Res. 1999, 14, 1330-1337.
  • a first embodiment of a first aspect of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising: a. a first compound, said first compound being cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof; and b. a second compound, said second compound being a selective androgen receptor modulator, or a pharmaceutically acceptable salt thereof.
  • a second embodiment of a first aspect of this invention is a pharmaceutical composition of the first embodiment of the first aspect additionally comprising a pharmaceutical carrier or diluent.
  • a third embodiment of a first aspect of this invention is a pharmaceutical composition of the first or second embodiments of the first aspect wherein said first compound is (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1- yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof.
  • a fourth embodiment of a first aspect of this invention is a pharmaceutical composition as set forth in any of the first through third embodiments of the first aspect wherein said first compound is (-)-cis-6- phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, D-tartrate salt.
  • a fifth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is cyproterone, chlormadinone, flutamide, hydroxyflutamide, bicalutamide, nilutamide, or spironolactone, or a pharmaceutically acceptable salt thereof.
  • a sixth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is of the Formula
  • R 1 is hydrogen
  • R 2 is chloro, cyano or trifluoromethyl
  • R 1 and R 2 are each fluoro
  • R 3 and R 4 are each independently (C r C 6 )alkyl, (C 3 -C 7 )cycloall ⁇ yl or (C 2 -C 6 )alkenyl; or R 3 and R 4 taken together with the nitrogen to which they are attached is
  • n 1 , 2 or 3;
  • R 5 is (CrC 6 )alkyl optionally substituted with hydroxy or (C r C 6 )alkoxy;
  • a seventh embodiment of a first aspect of this invention is a pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is:
  • An eighth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment wherein said second compound is 4-(2-(S)-ethyl-piperidin-1-yl)-2-trifluoromethyl- benzonitrile or 4-((R)-sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile, or a pharmaceutically acceptable salt thereof.
  • a first embodiment of a second aspect of this invention is a method of treating a condition that presents with low bone mass, treating a wasting disease, increasing muscle mass, increasing lean body mass, decreasing fat body mass, treating bone fracture and muscle damage, treating female sexual dysfunction, treating prostate hypertrophy or treating male sexual dysfunction in a mammal in need thereof, the method comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition according to the first embodiment of the first aspect of this invention.
  • a second embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein the mammal has a condition that presents with low bone mass.
  • a third embodiment of a second aspect of this invention is the method of the second embodiment of the second aspect wherein the condition that presents with low bone mass is osteoporosis, a bone defect, childhood idiopathic bone loss, alveolar bone loss, mandibular bone loss, bone fracture, osteotomy, periodontitis or prosthetic ingrowth.
  • a fourth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein concomitant bone fracture and muscle damage are treated.
  • a fifth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein muscle mass is increased, lean body mass is increased or fat body mass is decreased.
  • a sixth embodiment of a second aspect of this invention is the method of the fifth embodiment of the second aspect wherein the mammal is a livestock mammal.
  • a seventh embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein female sexual dysfunction is treated.
  • An eighth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein the therapeutically effective amount of the pharmaceutical composition is about 0.01 mg/kg/day to about 20 mg/kg/day.
  • the mammal is a human, companion animal or livestock animal.
  • the term "companion animal” refers to a household pet or other domesticated animal such as, but not limited to, cattle, sheep, ferrets, swine, horses, rabbits, goats, dogs, cats and the like.
  • the term "livestock mammal” refers to domesticated mammals such as, but not limited to, cattle, horses, llamas, goats, swine, sheep or rabbits.
  • the methods of this invention are also useful when the animal is a non-mammal such as a bird or fish. In all of the methods of this invention a most preferred mammal is a human.
  • compositions and methods of this invention result in higher magnitude bone mass gain than is achievable with the same doses of cis-6-phenyl-5-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]- 5,6,7,8-tetrahydro-napthalene-2-ol as described above alone or a selective androgen receptor modulator, such as 4-(2-(S)-ethyI-piperidin-1-yl)-2-trifluoromethyl-benzonitrile, alone.
  • compositions of this invention increase bone mass and will decrease fracture rates to a greater extent than is achievable through the use of either cis-6-phenyl-5-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-5,6,7,8- tetrahydro-napthalene-2-ol or the selective androgen receptor modulator alone.
  • This invention provides compositions and methods that are useful for increasing bone mass, increasing muscle mass, decreasing fat body mass and preventing the thinning of the vaginal luminal epithelium.
  • condition which presents with low bone mass refers to a condition where the level of bone mass is below the age specific normal as defined in standards by the World Health Organization "Assessment of Fracture Risk and its Application to Screening for Postmenopausal Osteoporosis (1994), Report of a World Health Organization Study Group. World Health Organization Technical Series 843". Childhood idiopathic and primary osteoporosis are also included. Included in the treatment of osteoporosis is the prevention or attenuation of long term complications such as curvature of the spine, loss of height, prosthetic surgery, and prevention of prostate malfunctioning. Also included is increasing the bone fracture healing rate and enhancing the rate of successful bone grafts. Also included is periodontal disease and alveolar bone loss.
  • condition which presents with low bone mass also refers to a mammal known to have a significantly higher than average chance of developing such diseases as are described above including osteoporosis (e.g. post-menopausal women, men over the age of 60, and persons being treated with drugs known to cause osteoporosis as a side-effect (such as glucocorticoid)).
  • osteoporosis e.g. post-menopausal women, men over the age of 60, and persons being treated with drugs known to cause osteoporosis as a side-effect (such as glucocorticoid)).
  • bone mass actually refers to bone mass per unit area which is sometimes (although not strictly correctly) referred to as bone mineral density.
  • musculoskeletal disorder refers to a condition wherein a subject has low bone mass and/or low muscle mass, and includes such diseases, disorders and conditions such as, but not limited to, conditions which present with low bone mass, osteoporosis, conditions which present with low muscle mass, muscle weakness, muscle atrophy, muscle wasting, osteotomy, childhood idiopathic bone loss, bone loss associated with periodontitis, bone healing following facial reconstruction, maxillary reconstruction, mandibular reconstruction and bone fracture. Further, musculoskeletal disorder encompasses such conditions as interfaces between newly attached prostheses and bone which require ingrowth.
  • Hypoactive sexual desire disorder is a disorder in which sexual fantasies and desire for sexual activity are persistently or recurrently diminished or absent, causing marked distress or interpersonal difficulties.
  • Hypoactive sexual desire disorder may be lifelong or acquired, generalized (global) or situational (partner-specific).
  • Sexual desire is a complex psychosomatic process based on brain activity (the "generator” or “motor” running in a rheostatic cyclic fashion), a poorly defined hormonal milieu, and cognitive scripting that includes sexual aspiration and motivation. Desynchronization of these components results in hypoactive sexual desire disorder.
  • hypoactive sexual desire disorder is commonly caused by boredom or unhappiness in a long-standing relationship, depression (which leads more often to decreased interest in sex than it does to impotence in the male or to inhibited excitement in the female), dependence on alcohol or psychoactive drugs, side effects from prescription drugs (eg, antihypertensives, antidepressants), and hormonal deficiencies.
  • This disorder can be secondary to impaired sexual functioning in the arousal or orgasm phase of the sexual response cycle.
  • Symptoms and signs of hypoactive sexual desire disorder include the patient complaining of a lack of interest in sex, even in ordinarily erotic situations.
  • the disorder is usually associated with infrequent sexual activity, often causing serious marital conflict.
  • Some patients have sexual encounters fairly often to please their partners and may have no difficulty with performance but continue to have sexual apathy.
  • boredom is the cause, frequency of sex with the usual partner decreases, but sexual desire may be normal or even intense with others (the situational form).
  • Sexual dysfunction due to a physical disorder is usually generalized (not specific to a given partner or situation). It is diagnosed when evidence from a patient's history, physical examination, or laboratory assessment can explain the dysfunction physiologically and when mental disorders that may better explain it can be ruled out. Resolution of the underlying physical disorders often results in resolution or amelioration of the sexual dysfunction.
  • the cause of sexual dysfunction is a combination of psychologic and physical factors, the appropriate diagnosis is sexual dysfunction due to combined factors. Sexual anhedonia (decreased or absent pleasure in sexual activity) is not an official diagnosis.
  • hypoactive sexual desire disorder Because loss of pleasure almost always results in loss of desire (although loss of desire may occur first).
  • the cause is likely to be depression or drugs if anhedonia is acquired and global (with all partners in all situations); interpersonal factors if anhedonia is confined to one partner or one situation; or repressive factors (eg, guilt, shame) due to family dysfunction or childhood trauma if anhedonia is lifelong.
  • sexual aversion is the probable diagnosis in lifelong cases.
  • Sexual arousal disorder is the persistent or recurrent inability to attain or to maintain the lubrication-swelling response of sexual excitement until completion of sexual activity. This disturbance occurs despite adequate focus, intensity, and duration of sexual stimulation.
  • the disorder may be lifelong or, more commonly, acquired and restricted to the partner.
  • the patient's complaints are usually related to lack of orgasm, although some women report lack of excitement.
  • the female sexual response phase of arousal is not easily distinguished from the phase of desire until physiological changes begin to take place in the vagina and clitoris as well as other sexual organs.
  • Vaginal engorgement enables transudation to occur and this process is responsible for increased vaginal lubrication. Transudation allows a flow of plasma through the epithelium and onto the vaginal surface, the driving force for which is increased blood flow in the vaginal capillary bed during the aroused state. In addition engorgement leads to an increase in vaginal length and luminal diameter, especially in the distal 2/3 of the vaginal canal. The luminal dilatation of the vagina is due to a combination of smooth muscle relaxation of its wall and skeletal muscle relaxation of the pelvic floor muscles. Some sexual pain ⁇ disorders such as vaginismus are thought to be due, at least in part, by inadequate relaxation preventing dilatation of the vagina; it has yet to be ascertained if this is primarily a smooth or skeletal muscle problem.
  • FSD Frequency Stimulation
  • Desire or libido is the drive for sexual expression. Its manifestations often include sexual thoughts either when in the company of an interested partner or when exposed to other erotic stimuli.
  • Arousal is the vascular response to sexual stimulation, an important component of which is vaginal lubrication and elongation of the vagina.
  • Orgasm is the release of sexual tension that has culminated during arousal.
  • FSD occurs when a woman has an inadequate or unsatisfactory response in any of these phases; desire, arousal or orgasm.
  • FSD categories include hypoactive sexual desire disorder, sexual arousal disorder, orgasmic disorders and sexual pain disorders.
  • Hypoactive sexual desire disorder is present if a woman has no or little desire to be sexual, and has no or few sexual thoughts or fantasies.
  • This type of FSD can be caused by low testosterone levels, due either to natural menopause or to surgical menopause. Other causes include illness, medications, fatigue, depression and anxiety.
  • FSAD sexual arousal disorder
  • the genitalia do not undergo the engorgement that characterizes normal sexual arousal.
  • the vaginal walls are poorly lubricated, so that intercourse is painful. Orgasms may be impeded.
  • Arousal disorder can be caused by reduced estrogen at menopause or after childbirth and during lactation, as well as by illnesses, with vascular components such as diabetes and atherosclerosis. Other causes result from treatment with diuretics, antihistamines, antidepressants, e.g., SSRIs or antihypertensive agents.
  • Sexual pain disorders (includes dyspareunia and vaginismus) is characterized by pain resulting from penetration and may be caused by medications which reduce lubrication, endometriosis, pelvic inflammatory disease, inflammatory bowel disease or urinary tract problems.
  • Dyspareunia is painful coitus or attempted coitus. Dyspareunia is usually introital but may also occur before, during, or after intercourse. Causes include menopausal involution with dryness and thinning of the mucosa. Pain during or after coitus is the chief complaint.
  • the term "treating”, “treat” or “treatment” as used herein includes curative, preventative (e.g. prophylactic) and palliative treatment.
  • the parenthetical negative or positive sign used herein denotes the direction a plane of polarized light is rotated by the particular stereoisomer.
  • the compound (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol rotates the plane of polarized light in a counterclockwise or (-) direction
  • (+)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6J,8-tetrahydronapthalene-2-ol rotates the plane of polarized light in a clockwise or (+) direction.
  • pharmaceutically acceptable is meant that the vehicle, carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • pharmaceutically-acceptable salt refers to nontoxic anionic salts containing anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate.
  • anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate.
  • nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl-glucamine), benethamine (N- benzylphenethylamine), piperazine or tromethamine (2-amino-2-hydroxymethyl-1 ,3-propanediol).
  • nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl-glucamine), benethamine (N- benzylphenethylamine), piperazine or tromethamine (2-amino-2-hydroxymethyl-1 ,3-
  • reaction-inert solvent and “inert solvent” refers to a solvent or a mixture of solvents that does not interact with starting materials, reagents, intermediates or products in a manner which adversely affects the yield of the desired product.
  • (C r C 6 )alkyl means a straight or branched alkyl group having from one to six carbons.
  • Examples of “(C r C 6 )alkyl” include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl and neopentyl.
  • (C 3 -C 7 )cycloalkyl means a cycloalkyl group having from three to seven carbons.
  • (C 3 -C 7 )cycloalkyl examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • (C 2 -C 6 )alkenyl means a straight or branched alkenyl group having from two to six carbons.
  • Examples of “(C 2 -C 6 )alkenyl” include, but are not limited to, vinyl, allyl, propenyl, butenyl, pentenyl, hexenyl, isopropenyl, isobutenyl, sec-butenyl and neopentenyl.
  • (C 1 - C 6 )alkoxy means a straight or branched alkoxy group having from one to six carbons.
  • Examples of “(Cr C 6 )alkoxy” include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy, isopropoxy, isobutoxy, sec-butoxy and neopentoxy.
  • the chemist of ordinary skill will recognize that the compounds used in the pharmaceutical compositions and methods of this invention may contain one or more atoms that may be in a particular stereochemical or geometric configuration, giving rise to stereoisomers and configurational isomers. All such isomers and mixtures thereof are included in this invention.
  • the first compound in the compositions of this invention is cis-6-phenyl-5-(4-(2-pyrrolidin-1-y!-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol, or a salt thereof.
  • the term cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)- 5,6,7, 8-tetrahydronapthalene-2-ol as used herein encompasses the individual (+) or (-) stereoisomers or any mixture thereof.
  • the first compound in the compositions is ( ⁇ -cis- ⁇ -phenyl-S ⁇ -pyrrolidin-i-yl-ethoxyJ-phenyO- ⁇ . ⁇ .Z. ⁇ -tetrahydronapthalene ⁇ -ol, and particularly the tartrate salt thereof. Hydrates and solvates of the compounds used in the compositions and methods of this invention are also included.
  • the compounds used in the compositions and methods of this invention can exist in isotopically labelled form, i.e., said compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • Isotopes of hydrogen, carbon, phosphorous, fluorine and chlorine include H, 2 H, 3 H, 12 C, 13 C, 14 C, 31 P, 32 P,
  • compositions and methods of this invention Compounds used in the compositions and methods of this invention, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug which contain those isotopes and/or other isotopes of other atoms are within the scope of this invention.
  • Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • certain deuterated, i.e., 2 H compounds may afford advantages with respect to metabolic stability and, hence, may be preferred.
  • Isotopically labelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • Other features and advantages of this invention will be apparent from this specification and the appendant claims that describe the invention.
  • Reaction Scheme I describes the synthesis of a compound of Formula I by a nucleophilic substitution reaction of the tertiary amine HNR 3 R 4 of formula III with an intermediate of formula II.
  • the group LG in the intermediate of formula Il represents any appropriate leaving group and typically a fluoro group is employed.
  • the reaction can be run neat or in an appropriate reaction-inert solvent. The reaction may be run at ambient temperature or with heating. The reaction is typically carried out neat between 65°C and 105 0 C using 1.5 to 2.5 equivalents of the amine HNR 3 R 4 for a period of 12 to 24 hours.
  • Certain of the 4-fluoro-benzonitrile derivatives, of formula Il wherein LG is fluoro, are known in the art and may be synthesized as described by Japanese Patent Application Number 01097937.
  • the resulting product, a compound of Formula I can be recovered by extraction, evaporation, or other techniques known in the art. It may then optionally be purified by chromatography, recrystallization, distillation, or other techniques known in the art. In certain cases the crude reaction mixture can be further reacted with another amine, such as 1 ,2-ethane-diamine, in order to consume any remaining starting material and facilitate the purification of the compound of Formula I.
  • Reaction Scheme 2 describes the synthesis of a compound of Formula I by a nucleophilic substitution reaction of the secondary amine H 2 NR 3 of Formula IV with an intermediate of formula Il to provide the intermediate of Formula V.
  • the nucleophilic substitution reaction employing the amine H 2 NR 3 and the intermediate of Formula Il can be carried out under the nucleophilic substitution conditions described above for Scheme 1.
  • the resulting product, an intermediate of Formula V can then be alkylated with an appropriate alkylating agent of formula R 4 X to provide the product of Formula I.
  • the group X in the alkylating agent R 4 X represents an appropriate leaving group, such as a halide and typically an iodide.
  • the alkylation reaction can be carried out in the presence of an appropriate base, such as sodium hydride or potassium hydride, in an appropriate aprotic solvent such as tetrahydrofuran (THF).
  • the alkylation reaction is typically carried out at ambient temperature for a period of one to twenty four hours by treating the intermediate of Formula V with two to three equivalents of an appropriate base in an appropriate solvent followed by addition of two equivalents of the alkylating agent R 4 X.
  • the reaction mixture can be quenched by addition of water and the product of Formula I can be recovered by extraction, evaporation, or other techniques known in the art. It may then optionally be purified by chromatography, recrystallization, distillation, or other techniques known in the art.
  • some of the methods useful for the preparation of such compounds may require protection of a particular functionality, e.g., to prevent interference by such functionality in reactions at other sites within the molecule or to preserve the integrity of such functionality.
  • protection is readily determined by one skilled in the art, and will vary depending on, for example, the nature of the functionality and the conditions of the selected preparation method. See, e.g., T.W. Greene, Protective Groups in Organic Synthesis. John Wiley & Sons, New York, 1991.
  • Some of the compounds used in the compositions and methods of this invention are acidic and they form a salt with a pharmaceutically acceptable cation.
  • Some of the compounds used in the compositions and methods of this invention are basic and they form a salt with a pharmaceutically acceptable anion. All such salts are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate.
  • the salts are recovered either by filtration, by precipitation with a non-solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate.
  • the compounds are obtained in crystalline form according to procedures known in the art, such as by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
  • the compounds of Formula I are selective androgen receptor modulators which have an affinity for the androgen receptor and cause a biological effect by binding to the receptor.
  • the compounds of Formula I act as agonists, which may exhibit tissue selective androgen receptor agonist activity.
  • the compounds of Formula I that exhibit androgen receptor agonist activity can be employed to treat conditions responsive to agonism of the androgen receptor. Examples of such conditions include, but are not limited to, conditions that present with low bone mass, such as osteoporosis, frailty, an osteoporotic fracture, a bone defect, childhood idiopathic bone loss, alveolar bone loss, mandibular bone loss, bone fracture, osteotomy, periodontitis or prosthetic ingrowth.
  • osteoporosis includes primary osteoporosis, such as senile, postmenopausal and juvenile osteoporosis, as well as secondary osteoporosis, such as osteoporosis due to hyperthyroidism or Gushing syndrome (due to corticosteroid use), acromegaly, hypogonadism, dysosteogenesis and hypophospatasemia.
  • the compounds of the invention with androgen receptor agonist activity may also be employed for treating wasting diseases (such as post operative, tumor, trauma, chronic renal disease or AIDS induced), male hypogonadism, male sexual dysfunction (male erectile dysfunction, male dysspermatogenic sterility), abnormal sex differentiation (male hermaphroditism), male delayed puberty, male infertility, aplastic anemia, hemolytic anemia, sickle cell anemia, renal anemia, idiopathic thrombocytopenic purpura, myelofibrosis, inoperable breast cancer or mastopathy.
  • the compounds of the invention with androgen receptor agonist activity may also be used to increase muscle mass, increase lean body mass, decrease fat body mass or treat concomitant bone fracture and muscle damage.
  • Certain of the compounds of Formula I may exhibit androgen receptor antagonist activity and may further exhibit tissue selective androgen receptor antagonist activity.
  • Compounds acting as androgen receptor antagonists may be used to treat hormone dependent cancers such as prostate carcinomas, benign prostatic hyperplasia, acne, hirsutism, excess sebum production, alopecia, hypertrichosis, precocious puberty, prostamegaly, virilization and polycystic ovary syndrome.
  • Administration of the pharmaceutical compositions of this invention can be via any method that delivers the composition systemically and/or locally. These methods include, but are not limited to, oral routes, parenteral, transdermal and intraduodenal routes, etc.
  • a pharmaceutical composition of this invention is administered orally, but parenteral administration (e.g., intravenous, intramuscular, subcutaneous or intramedullary) may be utilized, for example, where oral administration is inappropriate for the target or where the patient is unable to ingest the drug.
  • an effective dosage for cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol or a salt thereof, used in the pharmaceutical compositions and methods of this invention is in the range of 0.001 to 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day.
  • an effective dosage for the selective androgen receptor modulator, such as the Formula I compound or a salt thereof, used in the pharmaceutical compositions and methods of this invention is in the range of 0.001 to 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day.
  • a preferred therapeutically effective amount of the pharmaceutical composition of this invention is about 0.01 mg/kg/day to about 20 mg/kg/day.
  • 2-ol or a salt thereof and a selective androgen receptor modulator when used in the methods of the present invention is generally administered in the form of a pharmaceutical composition
  • a pharmaceutical composition comprising the combination together with a pharmaceutically acceptable vehicle, diluent or carrier.
  • the pharmaceutical composition of this invention can be administered in any conventional oral, parenteral, rectal or transdermal dosage form.
  • the pharmaceutical composition can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like.
  • Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often very useful for tabletting purposes.
  • Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • a preferred formulation is a solution or suspension in an oil, for example olive oil, Miglyol® (Condea Vista Co., Cranford, NJ) or Capmul® (Abitec Corporation, Colombus, OH), in a soft gelatin capsule.
  • Antioxidants may be added to prevent long term degradation as appropriate.
  • the compounds of this invention can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diluents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
  • solutions in sesame or peanut oil or in aqueous propylene glycol can be employed, as well as sterile aqueous solutions of the corresponding water-soluble salts.
  • aqueous solutions may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal injection purposes.
  • the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
  • dilute sterile, aqueous or partially aqueous solutions are prepared.
  • aqueous or partially aqueous solutions are prepared.
  • Methods of preparing various pharmaceutical compositions with a certain amount of active ingredient are known, or will be apparent in light of this disclosure, to those skilled in this art.
  • Remington's Pharmaceutical Sciences Mack Publishing Company, Easton, Pa., 19th Edition (1995).
  • compositions according to the invention may contain 0.1%-95% of the pharmaceutical combination or a salt thereof of this invention, preferably 1 %-70%.
  • the composition or formulation to be administered will contain a quantity of the pharmaceutical combination or a salt thereof according to the invention in an amount effective to treat the disease/condition of the subject being treated.
  • active ingredient means a combination of a first compound, cis-6- phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof and a second compound, a selective androgen receptor modulator.
  • the first compound of the "active ingredient" used in the formulation is (-)-cis-6-phenyl-5-(4- (2-pyrrolidin-1 -yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, tartrate salt.
  • Formulation 1 Gelatin Capsules
  • Hard gelatin capsules are prepared using the following:
  • a tablet formulation is prepared using the ingredients below: Formulation 2: Tablets
  • tablets each containing 0.25-100 mg of active ingredients are made up as follows:
  • Talc 1 The active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly.
  • the solution of polyvinylpyrrolidone is mixed with the resultant powders which are then passed through a No. 14 mesh U.S. sieve.
  • the granules so produced are dried at 50° - 60°C and passed through a No. 18 mesh U.S. sieve.
  • the sodium carboxymethyl starch, magnesium stearate, and talc previously passed through a No. 60 U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets.
  • Suspensions each containing 0.25-100 mg of active ingredient per 5 ml dose are made as follows: Formulation 4: Suspensions
  • the active ingredient is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form smooth paste.
  • the benzoic acid solution, flavor, and color are diluted with some of the water and added, with stirring. Sufficient water is then added to produce the required volume.
  • Aerosol solution is prepared containing the following ingredients: Formulation 5: Aerosol
  • Propellant 22 (Chlorodifluoromethane) 70.00
  • the active ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 3O 0 C, and transferred to a filling device.
  • the required amount is then fed to a stainless steel container and diluted with the remaining propellant.
  • the valve units are then fitted to the container.
  • Suppositories are prepared as follows: Formulation 6: Suppositories
  • the solution of the above ingredients is intravenously administered to a patient at a rate of about 1 ml. per minute.
  • Soft gelatin capsules are prepared using the following: Formulation 8: Soft Gelatin Capsule with Oil Formulation
  • (+)-2-ethyl-piperidine was dissolved in cooled H 2 O and solid potassium hydroxide was added to bring the pH of the solution to 14.
  • the (+)-2-ethyl-piperidine (15.88 g) was extracted with diethyl ether (3x), dried (MgSO 4 ), filtered, and concentrated to a clear oil. [ ⁇ ] 589 +4.88 o (0.413g/mL, CHCI 3 ).
  • (+)-2-Ethyl-piperidine (3.0 g, 26.5 mmol., Preparation 1) and 4-fluoro-2-trifluoromethyl-benzonitrile (2.0 g, 10.6 mmol.) were heated neat at 65°C overnight.
  • the reaction mixture was cooled and partitioned between diethyl ether and 1 N HCI.
  • the organic layers were combined, dried (MgSO 4 ), filtered, and evaporated to dryness.
  • the resulting residue (0.5 g, 2.64 mmol.) was a mixture of desired product and starting 4-fluoro-2-trifluoromethyl-benzonitrile.
  • Example 2 The general procedure described in Example 1 was followed by reacting 4-fluoro-2-trifluoromethyl- benzonitrile with 2-ethyl-piperidine at 70°C to yield the desired crude product.
  • the reaction mixture was cooled and partitioned between dichloromethane and 2M HCI.
  • the organic layer was dried (MgSO 4 ), filtered, and concentrated to dryness.
  • the residue was purified by preparative thin-layer chromatography (TLC) using 30% ethyl acetate/hexanes as the eluant to give the title compound.
  • Step B Preparation of 4-((R)-sec-Butyl-ethyl-amino)-2-chloro-benzonitrile
  • Examples 9, 10, 12, 13, 14 and 27 were prepared by following the general procedure of Example 8, Step B, but substituting appropriate benzonitrile derivative and alkyl iodide.
  • the compounds of Examples 15, 16, 17, 18, 19, 20 and 22-23 were prepared by following the general procedure of Example 8, Step A, but substituting the appropriate benzonitrile derivative and appropriate amine.
  • Examples 11, 21 and 24-26 were prepared by following the general procedure of Example 8, Steps A and B, but substituting the appropriate benzonitrile derivative, amine and alkyl iodide.
  • Step A Preparation of 4-(R)-sec-Butyl-propyl-amino)-phthalonitrile 4-Fluoro-phthalonitrile was reacted with (-)-sec-butylamine to afford the desired compound.
  • Step A Preparation of 4-(1-(R). 2-dimethyl-propylamino)-2-trifluoromethyl-benzonitrile 4-Fluoro-2-trifluoromethyl-benzonitrile was reacted with 1-(R), 2-dimethyl-propylamine at 90 0 C to afford the desired product.
  • Step B Preparation of 4-Fd-(R), 2-dimethyl-propyl)-methyl-aminol-2-trifluoromethyl-benzonitrile
  • Examples 30-53 Table 1 , below, provides Examples 30-53.
  • Examples 30 through 53 can be prepared by methods analogous to the methods employed for the preparation of Examples 1-29.
  • the compounds of Examples 30-53 can be purified and characterized according to methods analogous to the methods used for Examples 1-29.
  • All salts of the Formula I compound are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate.
  • the salts are recovered either by filtration, by precipitation with a non-solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate.
  • the compounds can be obtained in crystalline form by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
  • Formula I compound used in the compositions and methods of this invention, and the salts thereof are all adapted to therapeutic use as agents that mediate androgen receptors in mammals, particularly humans. By virtue of this activity, these agents are useful for treating conditions that present with low bone mass and improve frailty and other disease/conditions detailed above.
  • the utility of the Formula I compound of the invention and the salts thereof as medical agents in the treatment of the above described disease/conditions in mammals is demonstrated by the activity of the compound of this invention in conventional assays and the in vitro and in vivo assays described below.
  • the in vitro and in vivo assays may be used to determine the activity of analogous agents as well as the compounds of this invention.
  • Such assays also provide a means whereby the activities of the Formula I compound of this invention, and the salts thereof can be compared with the activities of other known compounds. The results of these comparisons are useful for determining dosage levels in mammals, including humans, for the treatment of such diseases.
  • Table 2 below, provides data obtained for compounds of the invention using the human androgen receptor binding analysis assay described hereinabove.
  • the purpose of this study is to test the effects of test compound in aged intact or ovariectomized (OVX) female rat model.
  • pQCT peripheral quantitative computerized tomography
  • volumetric total, trabecular and cortical bone mineral content and density were determined.
  • Peripheral Quantitative Computerized Tomography (pQCT) Analysis Excised femurs were scanned by a pQCT X-ray machine (Stratec XCT Research M, Norland Medical Systems, Fort Atkinson, Wl.) with software version 5.40.
  • a 1 millimeter (mm) thick cross section of the femur metaphysis was taken at 5.0 mm (proximal femoral metaphysis, a primary cancellous bone site) and 13 mm (femoral shafts, a cortical bone site) proximal from the distal end with a voxel size of 0.10 mm.
  • Cortical bone was defined and analyzed using contour mode 2 and cortical mode 4.
  • An outer threshold setting of 340 mg/cm 3 was used to distinguish the cortical shell from soft tissue and an inner threshold of 529 mg/cm 3 to distinguish cortical bone along the endocortical surface.
  • Trabecular bone was determined using peel mode 4 with a threshold of 655 mg/cm 3 to distinguish (sub)cortical from cancellous bone. An additional concentric peel of 1% of the defined cancellous bone was used to ensure that (sub)cortical bone was eliminated from the analysis. Volumetric content, density, and area were determined for both trabecular and cortical bone (Jamsa T. et al., Bone 23:155-161 , 1998; Ke, H.Z. et al., Journal of Bone and Mineral Research, 16:765-773, 2001). Using the above setting, it was determined that the ex vivo precision of volumetric content, density and area of total bone, trabecular, and cortical regions ranged from 0.99% to 3.49% with repositioning.
  • Vaginal histology Vaginal tissue was fixed and embedded in paraffin. Five micron sections were cut and stained with Alcian Blue staining. Histology examination of vaginal luminal epithelial thickness and mucopolysaccharide (secreted cells) was performed.
  • Group III OVX + Vehicle
  • Group IV OVX + SARM Test Compound at 10 mg/kg/day (in Vehicle)
  • Group Vl OVX + SARM Test Compound at 10 mg/kg/day and SERM Test Compound at 0.1 mg/kg/day.
  • the SERM compound employed in experimental groups V and Vl is cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,- tetrahydronaphthalene-2-ol.
  • the vehicle employed for the SERM test compound is one milliliter of 0.5% methylcellulose. The results are provided below in Table 3.
  • Table 3 The values provided in Table 3 are the mean values ⁇ the standard error measurement.
  • DFM distal femoral metaphysis
  • FS femoral shafts
  • g grams
  • mg/cm 3 milligrams per cubic centimeter
  • mg/mm milligrams per millimeter
  • a p ⁇ 0.05 vs. Baseline
  • b p ⁇ 0.05 vs. Sham
  • c p ⁇ 0.05 vs. OVX
  • d p ⁇ 0.05 vs.
  • SARM compound e: p ⁇ 0.05 vs.
  • SERM compound mg/kg/day: milligrams per kilogram of body weight per day; and ml: milliliter.
  • Body weight and body composition As shown in Table 3, OVX rats treated with 4-(2-(S)-ethyl- piperidin-1-yl)-2-trifluoromethyl-benzonitrile (SARM) had little effect on body weight, while cis-6-phenyl- 5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol (SERM) and the combination of SARM and SERM decreased body weight as compared with vehicle-treated OVX controls.
  • SARM 4-(2-(S)-ethyl- piperidin-1-yl)-2-trifluoromethyl-benzonitrile
  • Distal femoral pQCT analysis showed that total mineral density and trabecular mineral density of distal femoral metaphysis significantly decreased in vehicle- treated OVX group as compared to vehicle treated sham controls.
  • Total mineral density and trabecular mineral density of distal femoral metaphysis increased significantly in cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl- ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol (SERM) treated OVX rats or in the SERM and SARM combination group as compared with vehicle treated OVX controls. These two parameters did not differ between SARM treated OVX rats compared with vehicle treated OVX controls.
  • Femoral shaft pQCT analysis showed that total mineral content and cortical mineral content of distal femoral metaphysis significantly decreased in vehicle-treated OVX group as compared to vehicle treated sham controls.
  • Total mineral content and cortical mineral content of femoral shafts increased significantly in SARM treated OVX rats or in the SERM and SARM combination group as compared with vehicle treated OVX controls. These two parameters did not differ significantly between SERM treated OVX rats compared with vehicle treated OVX controls.
  • OVX caused dramatically thinning of vaginal luminal epithelial and decrease in mucopolysaccharide (secreted cells) staining.
  • Treatment with cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl- ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphtha-lene-2-ol (SERM) partially prevented these changes induced by OVX
  • SARM 4-(2-(S)-ethyI-piperidin-1-yl)-2-trifluoromethyl-benzonitrile
  • the purpose of this study is to test the effects of test compound in aged intaGt or orchidectomized (ORX) male rat model.
  • mice at 11 months of age were sham-operated or ORX.
  • the subcutaneous injections were given 2 times (Tuesday and Friday) per week with the first injection (Tuesday) at 9 or 30 mg/kg (in sesame oil, total 0.3 ml), and the second injection (Friday) at 12 or 40 mg/kg (in sesame oil, total 0.4 ml). All rats were injected subcutaneously with calcein at 10 mg/kg on 12 and 2 days before necropsy.
  • the experimental groups are as follows:
  • Treatment with the compound of Example 1 according to the above protocol decreased fat body mass and increased lean body mass in both sham and ORX rats in a dose dependant manner, with the exception of Group Il which showed a slight increase in fat body mass and slight decrease in lean body mass compared to Group I.
  • Treatment with the compound of Example 1 increased trabecular density in the distal femoral metaphysis (DFM) and total density in the femoral shaft (FS) in both sham and ORX rats.
  • DFM distal femoral metaphysis
  • FS femoral shaft
  • Treatment with the compound of Example 1 did not increase prostate weight in Group Il sham rats, although there was a slight increase in prostate weight of Group III compared to Group I. Treatment with the compound of Example 1 increased prostate weight in ORX rats to the level of sham controls in a dose dependent manner.
  • Fracture Technioue Sprage-Dawley rats at 3 months of age are anesthetized with Ketamine.
  • a 1 cm incision is made on the anteromedial aspect of the proximal part of the right tibia or femur.
  • Intramedullary nailing is performed with a 0.8 mm stainless steel tube (maximum load 36.3 N, maximum stiffness 61.8 N/mm, tested under the same conditions as the bones). No reaming of the medullary canal is performed.
  • a standardized closed fracture is produced 2 mm above the tibiofibular junction by three-point bending using specially designed adjustable forceps with blunt jaws. To minimize soft tissue damage, care is taken not to displace the fracture.
  • the skin is closed with monofilament nylon sutures. The operation is performed under sterile conditions. Radiographs of all fractures are taken immediately after nailing, and rats with fractures outside the specified diaphyseal area or with displaced nails are excluded. The remaining animals are divided randomly into the following groups with 10 - 12 animals per each subgroup per time point for testing the fracture healing.
  • 10 - 12 rats from each group are anesthetized with Ketamine and sacrificed by exsanguination. Both tibiofibular bones are removed by dissection and all soft tissue is stripped. Bones from 5 - 6 rats for each group are stored in 70% ethanol for histological analysis, and bones from another 5 - 6 rats for each group are stored in a buffered Ringer's solution (+4°C, pH 7.4) for radiographs and biomechanical testing which is performed.
  • Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site. The following measurements are performed: (1) fracture gap - measured as the shortest distance between the cortical bone ends in the fracture, (2) callus length and callus diameter, (3) total bone volume area of callus, (4) bony tissue per tissue area inside the callus area, (5) fibrous tissue in the callus, and (6) cartilage area in the callus.
  • Biomechanical Analysis The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue lnt 45:292-297, 1989). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedure. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.
  • the fracture site is sawed 3 cm to each side of the fracture line, embedded undecalcified in methymethacrylate, and cut on a Reichert-Jung Polycut microtome in 8 ⁇ m thick of frontal sections.
  • Masson-Trichrome stained mid-frontal sections (including both tibia and fibula) are used for visualization of the cellullar and tissue response to fracture healing with and without treatment. Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site.
  • fracture gap measured as the shortest distance between the cortical bone ends in the fracture
  • callus length and callus diameter (3) total bone volume area of callus
  • bony tissue per tissue area inside the callus area (4) bony tissue per tissue area inside the callus area
  • fibrous tissue in the callus (6) cartilage area in the callus.
  • Biomechanical Analysis The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue lnt 45:292-297, 1989) and Peter et al. (Peter, C.P.; Cook, W.O.; Nunamaker, D.M.; Provost, M. T.; Seedor, J.G.; Rodan, G.A. Effects of Alendronate On Fracture Healing And Bone Remodeling In Dogs. J. Orthop. Res. 14:74-70, 1996). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedures. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Epidemiology (AREA)
  • Neurology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Child & Adolescent Psychology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

This invention relates to a pharmaceutical combination of a selective estrogen receptor modulator (SERM) and a selective androgen receptor modulator (SARM). Particularly, this invention relates to a pharmaceutical composition comprising cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8­tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof; and a selective androgen receptor modulator. This invention also relates to methods of treatment using the pharmaceutical composition comprising cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene­2-ol, or a pharmaceutically acceptable salt thereof; and a selective androgen receptor modulator. Particularly, this invention is directed to methods to prevent and/or restore age-related decline in muscle mass and strength, treat a wasting disease, treat a condition that prevents with low bone mass, increase muscle mass, increase lean body mass, decrease fat body mass, treat bone fracture and muscle damage and treat female sexual dysfunction in mammals, including humans.

Description

THERAPEUTIC COMBINATIONS COMPRISING A SELECTIVE ESTROGEN RECEPTOR MODULATOR AND A SELECTIVE ANDROGEN RECEPTOR MODULATOR BACKGROUND OF THE INVENTION
The present invention relates to a pharmaceutical combination of a selective estrogen receptor modulator (SERM) and a selective androgen receptor modulator (SARM) that is useful for the prevention and treatment of musculoskeletal disorders and female sexual dysfunction. The present invention also relates to the use of such combinations to treat female sexual dysfunction and musculoskeletal disorders, including osteoporosis, osteoporotic fracture, low bone mass, frailty and the like in mammals, including humans. In particular, this invention relates to a combination of cis-6-phenyl-5-(4-(2-pyrrolidin- 1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol, or a pharmaceutically acceptable salt thereof and a selective androgen receptor modulator.
Osteoporosis is a systemic skeletal disease, characterized by low bone mass and deterioration of bone tissue with a consequent increase in bone fragility and susceptibility to fracture. In the U.S., the condition affects more than 25 million people and causes more than 1.3 million fractures each year, including 500,000 spine, 250,000 hip and 240,000 wrist fractures annually. Hip fractures are the most serious, and are associated with a 20% excess mortality in the year following fracture, and over 50% of the survivors being incapacitated. The elderly are at greatest risk of osteoporosis, and the problem is therefore expected to increase significantly during the next several decades with the aging of the population and by increasing longevity. The cost of managing fractures is substantial as approximately $13.8 billion dollars were spent in the U.S. in 1995 alone. Worldwide fracture incidence is forecast to increase three-fold over the next 60 years, and one study estimates that there will be 4.5 million hip fractures worldwide in 2050. The direct as well as indirect costs of fractures are therefore expected to increase correspondingly.
Although both men and women are susceptible to musculoskeletal disorders, including osteoporosis, women are at greater risk than men. Women experience a sharp acceleration of bone loss following menopause. The recent National Osteoporosis Risk Assessment, a study of 200,160 ambulatory postmenopausal women aged 50 years or older with no previous diagnosis of osteoporosis, using World Health Organization criteria, found that 39.6% had osteopenia and 7.2% had osteoporosis (Siris, E.S. et al., JAMA 2001, 286(22), 2815-2822). In the same study, age, personal or family history of fracture, Asian or Hispanic heritage, smoking, and cortisone use were associated with significantly increased likelihood of osteoporosis; whereas higher body mass index, African American heritage, estrogen or diuretic use, exercise, and alcohol consumption significantly decreased the likelihood. Estrogen is an agent useful for preventing osteoporosis or postmenopausal bone loss in women. In addition, Black, et al., in EP 0605193A1 report that estrogen, particularly when taken orally, lowers plasma levels of LDL and raises those of the beneficial high density lipoproteins (HDL's).
Recently, a number of selective estrogen receptor modulators have been proposed for treatment of osteoporosis. It has been reported (Osteoporosis Conference Scrip No. 1812/13 Apr. 16/20, 1993, p. 29) that raloxifene, 6-hydroxy-2-(4-hydroxyphenyl)-3-[4-(2-piperidinoethoxy)benzoyl]benzo[b]thiophene, mimics the favorable action of estrogens on bone and lipids but, unlike estrogen, has minimal uterine stimulatory effect. [Black, LJ. et al., Raloxifene (LY139481 HCI) Prevents Bone Loss and Reduces Serum Cholesterol Without Causing Uterine Hypertrophy in Ovariectomized Rats, J. Clin. Invest., 1994, 93, 63-69 and Delmas, P.D. et al., Effects of Raloxifene on Bone Mineral Density, Serum Cholesterol Concentration, and Uterine Endometrium in Postmenopausal Women, New England Journal of Medicine, 1997, 337, 1641-1647].
Also, tamoxifen, 1-(4-b-dimethylaminoethoxyphenyl)-1 ,2-diphenyl-but-1-ene, is an antiestrogen that is proposed as an osteoporosis agent which has a palliative effect on breast cancer, but is reported to have some estrogenic activity in the uterus.
U.S. Patent No. 5,254,595 discloses agents such as droloxifene, which prevent bone loss, reduce the risk of fracture and are useful for the treatment of osteoporosis.
Female sexual dysfunction (FSD) includes hypoactive sexual desire disorder, sexual anhedonia and dyspareunia. Proper sexual functioning in women depends on the sexual response cycle, which consists of an anticipatory mental set (sexual motive state or state of desire), effective vasocongestive arousal (swelling and lubrication), orgasm, and resolution. In women, orgasm is accompanied by contractions (not always subjectively experienced as such) of the muscles of the outer third of the vagina. Generalized muscular tension, perineal contractions, and involuntary pelvic thrusting (every 0.8 sec) usually occur. Orgasm is followed by resolution-a sense of general pleasure, well-being, and muscular relaxation. During this phase, women may be able to respond to additional stimulation almost immediately.
The sexual response cycle is mediated by a delicate, balanced interplay between the sympathetic and parasympathetic nervous systems. Vasocongestion is largely mediated by parasympathetic (cholinergic) outflow; orgasm is predominantly sympathetic (adrenergic). These responses are easily inhibited by cortical influences or by impaired hormonal, neural, or vascular mechanisms. Disorders of sexual response may involve one or more of the cycle's phases. Generally, both the subjective components of desire, arousal, and pleasure and the objective components of performance, vasocongestion, and orgasm are disturbed, although any may be affected independently. Sexual dysfunctions may be lifelong (no effective performance ever, generally due to intrapsychic conflicts) or acquired (after a period of normal function); generalized or limited to certain situations or certain partners; and total or partial.
The prevalence of FSD is difficult to gauge because the term covers several types of problems, some of which are difficult to measure, and because the interest in treating FSD is relatively recent. Many women's sexual problems are associated either directly with the female aging process or with chronic illnesses such as diabetes or hypertension.
There are wide variations in the reported incidence and prevalence of FSD, in part explained by the use of differing evaluation criteria, but most investigators report that a significant proportion of otherwise healthy women have symptoms of one or more of the FSD subgroups. By way of example, studies comparing sexual dysfunction in couples reveal that 63% of women had arousal or orgasmic dysfunction compared with 40% of men having erectile or ejaculatory dysfunction (Frank, E., Anderson, C. & Rubinstein, D., N. Enαl. J. Med.. 11 :229;111-115). However, the prevalence of female sexual arousal disorder varies considerably from survey to survey. In a recent National Health and Social Life Survey, 19% of women reported lubrication difficulties whereas 14% of women in an outpatient gynecological clinic reported similar difficulties with lubrication (Rosen, R., Taylor, J., Leiblum, S. et al., J. Sex Marital Then. 1993:19; 171-188).
Several studies have also reported dysfunction with sexual arousal in diabetic women (up to 47%), including reduced vaginal lubrication (Wincze, J. P., Albert, A. & Bansal, S., Arch. Sex Behav.. 1993:22; 587-601). There was no association between neuropathy and sexual dysfunction. Numerous studies have also shown that between 11-48% of women overall may have reduced sexual desire with age. Similarly, between 11-50% of women report problems with arousal and lubrication, and therefore experience pain with intercourse. Vaginismus is far less common, affecting approximately 1 % of women. Studies of sexually experienced women have detailed that 5-10% have primary anorgasmia. Another 10% have infrequent orgasms and a further 10% experience them inconsistently (Spector, IP. & Carey, M. P., Arch. Sex. Behav.. 1990:19; 389-408). FSAD is a highly prevalent sexual disorder affecting pre-, peri- and post menopausal (±HRT) women, it is associated with concomitant disorders such as depression, cardiovascular diseases, diabetes and UG disorders. The primary consequences of FSAD are lack of engorgement/swelling, lack of lubrication and lack of pleasurable genital sensation. The secondary consequences of FSAD are reduced sexual desire, pain during intercourse and difficulty in achieving an orgasm. It has recently been hypothesized that there is a vascular basis for at least a proportion of patients with symptoms of FSAD
(Goldstein, L. & Berman, J. R., Int. J. Impot. Res., 1998:10; S84-S90) with animal data supporting this view (Park, K., Goldstein, I., Andry, C, et al., Int. J. Impotence Res., 1997:9; 27-37).
The hormone estrogen has a profound effect in the vascular system of both men and women although its administration is associated with other effects that can be undesirable. Estrogen increases vasodilatation and inhibits the response of blood vessels to injury and the development of atherosclerosis. Estrogen-induced vasodilatation occurs 5 to 20 minutes after estrogen has been administered and is not dependent on changes in gene expression; this action of estrogen is sometimes referred to as "nongenomic." The estrogen-induced inhibition of the response to vascular injury and the preventive effect of estrogen against atherosclerosis occur over a period of hours or days after estrogen treatment and are dependent on changes in gene expression in the vascular tissues; these actions are sometimes referred to as "genomic."
U.S. Patent No. 5,552,412, incorporated herein by reference, discloses SERM compounds of the formula
Figure imgf000005_0001
wherein the variables are defined as set forth therein. Cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)- 5,6,7,8,-tetrahydronaphthalene-2-ol, ancj more particularly (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol is an orally active, highly potent SERM which prevents bone loss, decreases total serum cholesterol, and does not have estrogen-like uterine stimulating effects in OVX rats. U.S. Patent No. 5,948,809, also incorporated herein by reference, discloses (-)-cis-6-phenyl-5-(4-(2- pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol, tartrate salt. U.S. Patent Application No. 09/833,169, filed April 11 , 2001 , discloses methods of treating female sexual dysfunction with compounds such as cis-6-phenyl-5-(4-(2-pyrrolidin-1 -yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2- ol, of the Formula immediately hereinabove.
A selective androgen receptor modulator (SARM) is a compound that possesses androgenic activity and which exerts tissue-selective effects. SARM compounds can function as androgen receptor agonists, partial agonists, partial antagonists or antagonists. Examples of suitable SARMs include compounds such as cyproterone acetate, chlormadinone, flutamide, hydroxyflutamide, bicalutamide, nilutamide, spironolactone, 4-(trifluoromethyl)-2(1 H)-pyrrolidino[3,2-g] quinoline derivatives, 1 ,2-dihydropyridino [5,6- g]quinoline derivatives and piperidino[3,2-g]quinolinone derivatives.
Cypterone, also known as (ib^bJ-δ-chloro-i ^-dihydro-^-hydroxy-S'l-l-cyclopropaπ ^pregna- 1 ,4,6-triene-3,20-dione is disclosed in U.S. Patent 3,234,093. Chlormadinone, also known as 17- (acetyloxy)-6-chloropregna-4,6-diene-3,20-dione, in its acetate form, acts as an anti-androgen and is disclosed in U.S. Patent 3,485,852. Nilutamide, also known as 5,5-dimethyl-3-[4-nito-3-(trifluoromethyl) phenyl]-2,4-imidazolidinedione and by the trade name Nilandron® is disclosed in U.S. Patent 4,097,578. Flutamide, also known as 2-methyl-N-[4-nitro-3-(trifluoromethyl) phenyl] propanamide and the trade name Eulexin® is disclosed in U.S. Patent 3,847,988. Bicalutamide, also known as 4'-cyano-a',a',a'-trifluoro-3-(4- fluorophenylsulfonyl)-2-hydroxy-2-methylpropiono-m-toluidide and the trade name Casodex® is disclosed in EP-100172. The enantiomers of biclutamide are discussed by Tucker and Chesterton, J. Med. Chem. 1988, 31 , 885-887. Hydroxyflutamide, a known androgen receptor antagonist in most tissues, has been suggested to function as a SARM for effects on IL-6 production by osteoblasts as disclosed in Hofbauer et al. J. Bone Miner. Res. 1999, 14, 1330-1337. Additional SARMs have been disclosed in U.S. Patent 6,017,924; WO 01/16108, WO 01/16133, WO 01/16139, WO 02/00617, WO 02/16310, U.S. Patent Application Publication No. US 2002/0099096, U.S. Patent Application Publication No. US 2003/0022868, WO 03/011302 and WO 03/011824. SUMMARY OF THE INVENTION
A first embodiment of a first aspect of this invention is directed to a pharmaceutical composition comprising: a. a first compound, said first compound being cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof; and b. a second compound, said second compound being a selective androgen receptor modulator, or a pharmaceutically acceptable salt thereof.
A second embodiment of a first aspect of this invention is a pharmaceutical composition of the first embodiment of the first aspect additionally comprising a pharmaceutical carrier or diluent. A third embodiment of a first aspect of this invention is a pharmaceutical composition of the first or second embodiments of the first aspect wherein said first compound is (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1- yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof.
A fourth embodiment of a first aspect of this invention is a pharmaceutical composition as set forth in any of the first through third embodiments of the first aspect wherein said first compound is (-)-cis-6- phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, D-tartrate salt.
A fifth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is cyproterone, chlormadinone, flutamide, hydroxyflutamide, bicalutamide, nilutamide, or spironolactone, or a pharmaceutically acceptable salt thereof. A sixth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is of the Formula
Figure imgf000006_0001
a prodrug thereof or a pharmaceutically acceptable salt of said compound or said prodrug, wherein: R1 is hydrogen; R2 is chloro, cyano or trifluoromethyl; or R1 and R2 are each fluoro;
R3 and R4 are each independently (CrC6)alkyl, (C3-C7)cycloall<yl or (C2-C6)alkenyl; or R3 and R4 taken together with the nitrogen to which they are attached is
Figure imgf000006_0002
n is 1 , 2 or 3;
R5 is (CrC6)alkyl optionally substituted with hydroxy or (CrC6)alkoxy; and
R6 is hydrogen or (CrC6)alkyl optionally substituted with hydroxy or (CrC6)alkoxy. A seventh embodiment of a first aspect of this invention is a pharmaceutical composition of the first embodiment of the first aspect wherein said second compound is:
4-(2-ethyl-piperidin-1-yi)-2-trifluoromethyl-benzonitrile;
4-(2-ethyl-piperidin-1-yl)-2,6-difluoro-benzonitrile; 2-chloro-4-(2-ethyl-piperidin-1-yl)-benzonitrile;
4-(2-ethyl-piperidin-1-yl)-phthalonitrile;
4-(sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(sec-butyl-ethyl-amino)-2-chloro-benzonitrile;
4-(sec-butyl-methyl-amino)-2-chloro-benzonitrile; 4-(sec-butyl-propyl-amino)-2-chloro-benzonitrile;
4-(sec-butyl-propyl-amino)-phthalonitrile;
4-(sec-butyl-ethyl-amino)-2-trifluoromethyi-benzonitrile;
4-(sec-butyl-methyl-amino)-phthalonitrile;
4-(sec-butyl-ethyl-amino)-phthalonitrile; 4-dipropylamino-2-trifIuoromethyl-benzonitrile;
4-(ethyl-isopropyl-amino)-phthalonitrile;
4-dipropylamino-phthalonitrile;
4-diethylamino-2-trifluoromethyl-benzonitrile;
2-chloro-4-(ethyl-propyl-amino)-benzonitrile; 2-chloro-4-(isopropyl-methyl-amino)-benzonitrile;
4-[(1 ,2-dimethyl-propyl)-methyl-amino]-2-trifluoromethyl-benzonitrile;
2-chloro-4-[(1,2-dimethyl-propyl)-methyl-amino]-benzonitrile;
4-(2-methyl-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(cyclopentyl-methyl-amino)-2-trifluoromethyl-benzonitrile; 4-(2-propyl-piperidin-1 -yl)-2-trif luoromethyl-benzonitrile;
4-(2-ethyl-4-hydroxymethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(sec-butyl-propyI-amino)-2-trifluoromethyl-benzonitrile;
4-(allyl-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(2-methyl-piperidin-1-yI)-2-trifluoromethyi-benzonitrile; 4-(2-(ethoxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-(2-hydroxyethyl)-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-(methoxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(ethyl-pentyl-amino)-2-trifluoromethyl-benzonitrile;
4-(butyl-propyl-amino)-2-trifluoromethyl-benzonitrile; 4-(isopropyl-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(pentyl-propyl-amino)-2-trifluoromethyl-benzonitrile;
4-(2-hydroxymethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile;
4-(dibutyl-amino)-2-trifluoromethyl-benzonitrile; 4-(2-(hydroxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile; 2-chloro-4-(isopropyl-propyl-amino)-benzonitrile;
2-chloro-4-(diethyl-amino)-benzonitriie;
2-chloro-4-(ethyl-isopropyl-amino)-benzonitrile;
2-chloro-4-(dipropyl-amino)-benzonitriie; 4-(isopropyl-methyl-amino)-phthalonitrile;
4-(2-methoxymethyl-pyrrolidin-1-yl)-phthalonitrile;
4-azepan-1-yl-phthalonitrile; or
2,6-difluoro-4-(dipropyl-amino)-benzonitrile; or a stereoisomer thereof or a pharmaceutically acceptable salt of said compound or stereoisomer. An eighth embodiment of a first aspect of this invention is the pharmaceutical composition of the first embodiment wherein said second compound is 4-(2-(S)-ethyl-piperidin-1-yl)-2-trifluoromethyl- benzonitrile or 4-((R)-sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile, or a pharmaceutically acceptable salt thereof.
A first embodiment of a second aspect of this invention is a method of treating a condition that presents with low bone mass, treating a wasting disease, increasing muscle mass, increasing lean body mass, decreasing fat body mass, treating bone fracture and muscle damage, treating female sexual dysfunction, treating prostate hypertrophy or treating male sexual dysfunction in a mammal in need thereof, the method comprising administering to said mammal a therapeutically effective amount of a pharmaceutical composition according to the first embodiment of the first aspect of this invention. A second embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein the mammal has a condition that presents with low bone mass.
A third embodiment of a second aspect of this invention is the method of the second embodiment of the second aspect wherein the condition that presents with low bone mass is osteoporosis, a bone defect, childhood idiopathic bone loss, alveolar bone loss, mandibular bone loss, bone fracture, osteotomy, periodontitis or prosthetic ingrowth.
A fourth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein concomitant bone fracture and muscle damage are treated.
A fifth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein muscle mass is increased, lean body mass is increased or fat body mass is decreased.
A sixth embodiment of a second aspect of this invention is the method of the fifth embodiment of the second aspect wherein the mammal is a livestock mammal.
A seventh embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein female sexual dysfunction is treated. An eighth embodiment of a second aspect of this invention is the method of the first embodiment of the second aspect wherein the therapeutically effective amount of the pharmaceutical composition is about 0.01 mg/kg/day to about 20 mg/kg/day.
In all of the methods of this invention, it is preferred that the mammal is a human, companion animal or livestock animal. The term "companion animal" refers to a household pet or other domesticated animal such as, but not limited to, cattle, sheep, ferrets, swine, horses, rabbits, goats, dogs, cats and the like. The term "livestock mammal" refers to domesticated mammals such as, but not limited to, cattle, horses, llamas, goats, swine, sheep or rabbits. The methods of this invention are also useful when the animal is a non-mammal such as a bird or fish. In all of the methods of this invention a most preferred mammal is a human. The pharmaceutical compositions and methods of this invention result in higher magnitude bone mass gain than is achievable with the same doses of cis-6-phenyl-5-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]- 5,6,7,8-tetrahydro-napthalene-2-ol as described above alone or a selective androgen receptor modulator, such as 4-(2-(S)-ethyI-piperidin-1-yl)-2-trifluoromethyl-benzonitrile, alone. Thus, the pharmaceutical compositions of this invention increase bone mass and will decrease fracture rates to a greater extent than is achievable through the use of either cis-6-phenyl-5-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-5,6,7,8- tetrahydro-napthalene-2-ol or the selective androgen receptor modulator alone. This invention provides compositions and methods that are useful for increasing bone mass, increasing muscle mass, decreasing fat body mass and preventing the thinning of the vaginal luminal epithelium. Particularly, it has been found that the combination of cis-6-phenyl-5-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-5,6,7,8-tetrahydro-napthalene- 2-ol and a selective androgen receptor modulator (SARM), such as 4-(2-(S)-ethyl-piperidin-1-yl)-2- trifluoromethyl-benzonitrile, increases bone mass, increases muscle mass, decreases fat body mass and prevents the thinning of the vaginal luminal epithelium in mammals.
DETAILED DESCRIPTION OF THE INVENTION The phrase "condition which presents with low bone mass" refers to a condition where the level of bone mass is below the age specific normal as defined in standards by the World Health Organization "Assessment of Fracture Risk and its Application to Screening for Postmenopausal Osteoporosis (1994), Report of a World Health Organization Study Group. World Health Organization Technical Series 843". Childhood idiopathic and primary osteoporosis are also included. Included in the treatment of osteoporosis is the prevention or attenuation of long term complications such as curvature of the spine, loss of height, prosthetic surgery, and prevention of prostate malfunctioning. Also included is increasing the bone fracture healing rate and enhancing the rate of successful bone grafts. Also included is periodontal disease and alveolar bone loss.
The phrase "condition which presents with low bone mass" also refers to a mammal known to have a significantly higher than average chance of developing such diseases as are described above including osteoporosis (e.g. post-menopausal women, men over the age of 60, and persons being treated with drugs known to cause osteoporosis as a side-effect (such as glucocorticoid)).
Those skilled in the art will recognize that the term bone mass actually refers to bone mass per unit area which is sometimes (although not strictly correctly) referred to as bone mineral density.
The phrase "musculoskeletal disorder" refers to a condition wherein a subject has low bone mass and/or low muscle mass, and includes such diseases, disorders and conditions such as, but not limited to, conditions which present with low bone mass, osteoporosis, conditions which present with low muscle mass, muscle weakness, muscle atrophy, muscle wasting, osteotomy, childhood idiopathic bone loss, bone loss associated with periodontitis, bone healing following facial reconstruction, maxillary reconstruction, mandibular reconstruction and bone fracture. Further, musculoskeletal disorder encompasses such conditions as interfaces between newly attached prostheses and bone which require ingrowth.
Hypoactive sexual desire disorder is a disorder in which sexual fantasies and desire for sexual activity are persistently or recurrently diminished or absent, causing marked distress or interpersonal difficulties. Hypoactive sexual desire disorder may be lifelong or acquired, generalized (global) or situational (partner-specific). Sexual desire is a complex psychosomatic process based on brain activity (the "generator" or "motor" running in a rheostatic cyclic fashion), a poorly defined hormonal milieu, and cognitive scripting that includes sexual aspiration and motivation. Desynchronization of these components results in hypoactive sexual desire disorder. The acquired form of hypoactive sexual desire disorder is commonly caused by boredom or unhappiness in a long-standing relationship, depression (which leads more often to decreased interest in sex than it does to impotence in the male or to inhibited excitement in the female), dependence on alcohol or psychoactive drugs, side effects from prescription drugs (eg, antihypertensives, antidepressants), and hormonal deficiencies. This disorder can be secondary to impaired sexual functioning in the arousal or orgasm phase of the sexual response cycle.
Symptoms and signs of hypoactive sexual desire disorder include the patient complaining of a lack of interest in sex, even in ordinarily erotic situations. The disorder is usually associated with infrequent sexual activity, often causing serious marital conflict. Some patients have sexual encounters fairly often to please their partners and may have no difficulty with performance but continue to have sexual apathy. When boredom is the cause, frequency of sex with the usual partner decreases, but sexual desire may be normal or even intense with others (the situational form).
Clinically significant sexual dysfunction that causes personal distress or interpersonal problems and is most likely fully explained by direct physiologic effects of a physical disorder. Sexual dysfunction due to a physical disorder is usually generalized (not specific to a given partner or situation). It is diagnosed when evidence from a patient's history, physical examination, or laboratory assessment can explain the dysfunction physiologically and when mental disorders that may better explain it can be ruled out. Resolution of the underlying physical disorders often results in resolution or amelioration of the sexual dysfunction. When the cause of sexual dysfunction is a combination of psychologic and physical factors, the appropriate diagnosis is sexual dysfunction due to combined factors. Sexual anhedonia (decreased or absent pleasure in sexual activity) is not an official diagnosis. It is almost always classified under hypoactive sexual desire disorder, because loss of pleasure almost always results in loss of desire (although loss of desire may occur first). The cause is likely to be depression or drugs if anhedonia is acquired and global (with all partners in all situations); interpersonal factors if anhedonia is confined to one partner or one situation; or repressive factors (eg, guilt, shame) due to family dysfunction or childhood trauma if anhedonia is lifelong. Sexual aversion is the probable diagnosis in lifelong cases.
Sexual arousal disorder is the persistent or recurrent inability to attain or to maintain the lubrication-swelling response of sexual excitement until completion of sexual activity. This disturbance occurs despite adequate focus, intensity, and duration of sexual stimulation. The disorder may be lifelong or, more commonly, acquired and restricted to the partner. The patient's complaints are usually related to lack of orgasm, although some women report lack of excitement.
Although women can be orgasmic throughout their lives, sexual activity often decreases after age
60 because of the relative lack of partners and untreated physiologic changes (eg, atrophy of the vaginal mucosa, with resultant dryness and painful coitus).
The female sexual response phase of arousal is not easily distinguished from the phase of desire until physiological changes begin to take place in the vagina and clitoris as well as other sexual organs.
Sexual excitement and pleasure are accompanied by a combination of vascular and neuromuscular events which lead to engorgement of the clitoris, labia and vaginal wall, increased vaginal lubrication and dilatation of the vaginal lumen (Levin, RJ., Clin. Obstet. Gynecol.. 1980:7; 213-252; Ottesen, B.,
Gerstenberg, T., Ulrichsen, H. et al., Eur. J. Clin. Invest., 1983:13; 321-324; Levin, R.J.. Exp. Clin.
Endocrinol.. 1991 :98; 61-69; Levin, RJ. , Ann. Rev. Sex Res.. 1992:3; 1-48; Masters, W. H., Johnson, V. E.
Human Sexual Response. Little, Brown: Boston, 1996; Berman, J. R., Berman, L. & Goldstein, L., Urology.
1999:54; 385-391). Vaginal engorgement enables transudation to occur and this process is responsible for increased vaginal lubrication. Transudation allows a flow of plasma through the epithelium and onto the vaginal surface, the driving force for which is increased blood flow in the vaginal capillary bed during the aroused state. In addition engorgement leads to an increase in vaginal length and luminal diameter, especially in the distal 2/3 of the vaginal canal. The luminal dilatation of the vagina is due to a combination of smooth muscle relaxation of its wall and skeletal muscle relaxation of the pelvic floor muscles. Some sexual pain disorders such as vaginismus are thought to be due, at least in part, by inadequate relaxation preventing dilatation of the vagina; it has yet to be ascertained if this is primarily a smooth or skeletal muscle problem.
(Masters, W. H., Johnson, V. E. Human Sexual Response. Little, Brown: Boston, 1996; Berman, J. R.,
Berman, L. & Goldstein, L., Urology, 1999:54; 385-391). The categories of FSD are best defined by contrasting them to the phases of normal female sexual response: desire, arousal and orgasm. Desire or libido is the drive for sexual expression. Its manifestations often include sexual thoughts either when in the company of an interested partner or when exposed to other erotic stimuli. Arousal is the vascular response to sexual stimulation, an important component of which is vaginal lubrication and elongation of the vagina. Orgasm is the release of sexual tension that has culminated during arousal.
Hence, FSD occurs when a woman has an inadequate or unsatisfactory response in any of these phases; desire, arousal or orgasm. FSD categories include hypoactive sexual desire disorder, sexual arousal disorder, orgasmic disorders and sexual pain disorders.
Hypoactive sexual desire disorder is present if a woman has no or little desire to be sexual, and has no or few sexual thoughts or fantasies. This type of FSD can be caused by low testosterone levels, due either to natural menopause or to surgical menopause. Other causes include illness, medications, fatigue, depression and anxiety.
Sexual arousal disorder (FSAD) is characterized by inadequate genital response to sexual stimulation. The genitalia do not undergo the engorgement that characterizes normal sexual arousal. The vaginal walls are poorly lubricated, so that intercourse is painful. Orgasms may be impeded. Arousal disorder can be caused by reduced estrogen at menopause or after childbirth and during lactation, as well as by illnesses, with vascular components such as diabetes and atherosclerosis. Other causes result from treatment with diuretics, antihistamines, antidepressants, e.g., SSRIs or antihypertensive agents.
Sexual pain disorders (includes dyspareunia and vaginismus) is characterized by pain resulting from penetration and may be caused by medications which reduce lubrication, endometriosis, pelvic inflammatory disease, inflammatory bowel disease or urinary tract problems.
Dyspareunia is painful coitus or attempted coitus. Dyspareunia is usually introital but may also occur before, during, or after intercourse. Causes include menopausal involution with dryness and thinning of the mucosa. Pain during or after coitus is the chief complaint. The term "treating", "treat" or "treatment" as used herein includes curative, preventative (e.g. prophylactic) and palliative treatment.
The parenthetical negative or positive sign used herein (e.g., when found in parentheses in the name of a compound) denotes the direction a plane of polarized light is rotated by the particular stereoisomer. For example, the compound (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol rotates the plane of polarized light in a counterclockwise or (-) direction whereas (+)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6J,8-tetrahydronapthalene-2-ol rotates the plane of polarized light in a clockwise or (+) direction.
By "pharmaceutically acceptable" is meant that the vehicle, carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
The expression "pharmaceutically-acceptable salt" refers to nontoxic anionic salts containing anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate. The expression also refers to nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl-glucamine), benethamine (N- benzylphenethylamine), piperazine or tromethamine (2-amino-2-hydroxymethyl-1 ,3-propanediol).
As used herein, the expressions "reaction-inert solvent" and "inert solvent" refers to a solvent or a mixture of solvents that does not interact with starting materials, reagents, intermediates or products in a manner which adversely affects the yield of the desired product.
The term "(CrC6)alkyl" means a straight or branched alkyl group having from one to six carbons. Examples of "(CrC6)alkyl" include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl and neopentyl. The term "(C3-C7)cycloalkyl" means a cycloalkyl group having from three to seven carbons. Examples of "(C3-C7)cycloalkyl" include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. The term "(C2-C6)alkenyl" means a straight or branched alkenyl group having from two to six carbons. Examples of "(C2-C6)alkenyl" include, but are not limited to, vinyl, allyl, propenyl, butenyl, pentenyl, hexenyl, isopropenyl, isobutenyl, sec-butenyl and neopentenyl. The term "(C1- C6)alkoxy" means a straight or branched alkoxy group having from one to six carbons. Examples of "(Cr C6)alkoxy" include, but are not limited to, methoxy, ethoxy, propoxy, butoxy, pentoxy, hexoxy, isopropoxy, isobutoxy, sec-butoxy and neopentoxy. The chemist of ordinary skill will recognize that the compounds used in the pharmaceutical compositions and methods of this invention may contain one or more atoms that may be in a particular stereochemical or geometric configuration, giving rise to stereoisomers and configurational isomers. All such isomers and mixtures thereof are included in this invention. For example, the first compound in the compositions of this invention is cis-6-phenyl-5-(4-(2-pyrrolidin-1-y!-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol, or a salt thereof. The term cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)- 5,6,7, 8-tetrahydronapthalene-2-ol as used herein encompasses the individual (+) or (-) stereoisomers or any mixture thereof. In a preferred embodiment of this invention, the first compound in the compositions is (^-cis-θ-phenyl-S^^-pyrrolidin-i-yl-ethoxyJ-phenyO-δ.θ.Z.δ-tetrahydronapthalene^-ol, and particularly the tartrate salt thereof. Hydrates and solvates of the compounds used in the compositions and methods of this invention are also included.
It will be recognized that the compounds used in the compositions and methods of this invention can exist in isotopically labelled form, i.e., said compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Isotopes of hydrogen, carbon, phosphorous, fluorine and chlorine include H, 2H, 3H, 12C, 13C, 14C, 31P, 32P,
S, S, F, F, Cl and CI, respectively. Compounds used in the compositions and methods of this invention, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug which contain those isotopes and/or other isotopes of other atoms are within the scope of this invention. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, certain deuterated, i.e., 2H, compounds may afford advantages with respect to metabolic stability and, hence, may be preferred. Isotopically labelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent. Other features and advantages of this invention will be apparent from this specification and the appendant claims that describe the invention.
Certain processes for the manufacture of the compounds used in the pharmaceutical compositions and methods of this invention are provided below and are illustrated by the following description and by the examples. The compounds of Formula I as shown below are prepared as described in co-pending U.S. Provisional Patent Application No. 60/569,874, filed May 11 , 2004, the procedures of which are reproduced hereinbelow.
Synthesis
The compounds of Formula I can be prepared by methods analogously known in the art. One method for preparing these compounds is described below in Reaction Scheme I. Reaction Scheme I describes the synthesis of a compound of Formula I by a nucleophilic substitution reaction of the tertiary amine HNR3R4 of formula III with an intermediate of formula II. The group LG in the intermediate of formula Il represents any appropriate leaving group and typically a fluoro group is employed. The reaction can be run neat or in an appropriate reaction-inert solvent. The reaction may be run at ambient temperature or with heating. The reaction is typically carried out neat between 65°C and 1050C using 1.5 to 2.5 equivalents of the amine HNR3R4 for a period of 12 to 24 hours. Certain of the 4-fluoro-benzonitrile derivatives, of formula Il wherein LG is fluoro, are known in the art and may be synthesized as described by Japanese Patent Application Number 01097937. The resulting product, a compound of Formula I, can be recovered by extraction, evaporation, or other techniques known in the art. It may then optionally be purified by chromatography, recrystallization, distillation, or other techniques known in the art. In certain cases the crude reaction mixture can be further reacted with another amine, such as 1 ,2-ethane-diamine, in order to consume any remaining starting material and facilitate the purification of the compound of Formula I.
Reaction Scheme 1
Figure imgf000014_0001
Reaction Scheme 2 describes the synthesis of a compound of Formula I by a nucleophilic substitution reaction of the secondary amine H2NR3 of Formula IV with an intermediate of formula Il to provide the intermediate of Formula V. The nucleophilic substitution reaction employing the amine H2NR3 and the intermediate of Formula Il can be carried out under the nucleophilic substitution conditions described above for Scheme 1. The resulting product, an intermediate of Formula V, can then be alkylated with an appropriate alkylating agent of formula R4X to provide the product of Formula I. The group X in the alkylating agent R4X represents an appropriate leaving group, such as a halide and typically an iodide. The alkylation reaction can be carried out in the presence of an appropriate base, such as sodium hydride or potassium hydride, in an appropriate aprotic solvent such as tetrahydrofuran (THF). The alkylation reaction is typically carried out at ambient temperature for a period of one to twenty four hours by treating the intermediate of Formula V with two to three equivalents of an appropriate base in an appropriate solvent followed by addition of two equivalents of the alkylating agent R4X. The reaction mixture can be quenched by addition of water and the product of Formula I can be recovered by extraction, evaporation, or other techniques known in the art. It may then optionally be purified by chromatography, recrystallization, distillation, or other techniques known in the art. Reaction Scheme 2
Figure imgf000015_0001
As would be appreciated by those skilled in the art, some of the methods useful for the preparation of such compounds, as discussed above, may require protection of a particular functionality, e.g., to prevent interference by such functionality in reactions at other sites within the molecule or to preserve the integrity of such functionality. The need for, and type of, such protection is readily determined by one skilled in the art, and will vary depending on, for example, the nature of the functionality and the conditions of the selected preparation method. See, e.g., T.W. Greene, Protective Groups in Organic Synthesis. John Wiley & Sons, New York, 1991.
Some of the compounds used in the compositions and methods of this invention are acidic and they form a salt with a pharmaceutically acceptable cation. Some of the compounds used in the compositions and methods of this invention are basic and they form a salt with a pharmaceutically acceptable anion. All such salts are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate. The salts are recovered either by filtration, by precipitation with a non-solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate. The compounds are obtained in crystalline form according to procedures known in the art, such as by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
The compounds of Formula I are selective androgen receptor modulators which have an affinity for the androgen receptor and cause a biological effect by binding to the receptor. Typically, the compounds of Formula I act as agonists, which may exhibit tissue selective androgen receptor agonist activity. The compounds of Formula I that exhibit androgen receptor agonist activity can be employed to treat conditions responsive to agonism of the androgen receptor. Examples of such conditions include, but are not limited to, conditions that present with low bone mass, such as osteoporosis, frailty, an osteoporotic fracture, a bone defect, childhood idiopathic bone loss, alveolar bone loss, mandibular bone loss, bone fracture, osteotomy, periodontitis or prosthetic ingrowth. The term osteoporosis includes primary osteoporosis, such as senile, postmenopausal and juvenile osteoporosis, as well as secondary osteoporosis, such as osteoporosis due to hyperthyroidism or Gushing syndrome (due to corticosteroid use), acromegaly, hypogonadism, dysosteogenesis and hypophospatasemia. The compounds of the invention with androgen receptor agonist activity may also be employed for treating wasting diseases (such as post operative, tumor, trauma, chronic renal disease or AIDS induced), male hypogonadism, male sexual dysfunction (male erectile dysfunction, male dysspermatogenic sterility), abnormal sex differentiation (male hermaphroditism), male delayed puberty, male infertility, aplastic anemia, hemolytic anemia, sickle cell anemia, renal anemia, idiopathic thrombocytopenic purpura, myelofibrosis, inoperable breast cancer or mastopathy. The compounds of the invention with androgen receptor agonist activity may also be used to increase muscle mass, increase lean body mass, decrease fat body mass or treat concomitant bone fracture and muscle damage.
Certain of the compounds of Formula I may exhibit androgen receptor antagonist activity and may further exhibit tissue selective androgen receptor antagonist activity. Compounds acting as androgen receptor antagonists may be used to treat hormone dependent cancers such as prostate carcinomas, benign prostatic hyperplasia, acne, hirsutism, excess sebum production, alopecia, hypertrichosis, precocious puberty, prostamegaly, virilization and polycystic ovary syndrome.
Administration of the pharmaceutical compositions of this invention can be via any method that delivers the composition systemically and/or locally. These methods include, but are not limited to, oral routes, parenteral, transdermal and intraduodenal routes, etc. Generally, a pharmaceutical composition of this invention is administered orally, but parenteral administration (e.g., intravenous, intramuscular, subcutaneous or intramedullary) may be utilized, for example, where oral administration is inappropriate for the target or where the patient is unable to ingest the drug. in general an effective dosage for cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8- tetrahydronapthalene-2-ol or a salt thereof, used in the pharmaceutical compositions and methods of this invention is in the range of 0.001 to 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day. In general an effective dosage for the selective androgen receptor modulator, such as the Formula I compound or a salt thereof, used in the pharmaceutical compositions and methods of this invention is in the range of 0.001 to 100 mg/kg/day, preferably 0.01 to 10 mg/kg/day. A preferred therapeutically effective amount of the pharmaceutical composition of this invention is about 0.01 mg/kg/day to about 20 mg/kg/day. A combination of cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-
2-ol or a salt thereof and a selective androgen receptor modulator when used in the methods of the present invention is generally administered in the form of a pharmaceutical composition comprising the combination together with a pharmaceutically acceptable vehicle, diluent or carrier. Thus, the pharmaceutical composition of this invention can be administered in any conventional oral, parenteral, rectal or transdermal dosage form.
For oral administration the pharmaceutical composition can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like. Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often very useful for tabletting purposes. Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols. A preferred formulation is a solution or suspension in an oil, for example olive oil, Miglyol® (Condea Vista Co., Cranford, NJ) or Capmul® (Abitec Corporation, Colombus, OH), in a soft gelatin capsule. Antioxidants may be added to prevent long term degradation as appropriate. When aqueous suspensions and/or elixirs are desired for oral administration, the compounds of this invention can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diluents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
For purposes of parenteral administration, solutions in sesame or peanut oil or in aqueous propylene glycol can be employed, as well as sterile aqueous solutions of the corresponding water-soluble salts. Such aqueous solutions may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal injection purposes. In this connection, the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
For purposes of transdermal (e.g., topical) administration, dilute sterile, aqueous or partially aqueous solutions (usually in about 0.1% to 5% concentration), otherwise similar to the above parenteral solutions, are prepared. Methods of preparing various pharmaceutical compositions with a certain amount of active ingredient are known, or will be apparent in light of this disclosure, to those skilled in this art. For examples of methods of preparing pharmaceutical compositions, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 19th Edition (1995).
Pharmaceutical compositions according to the invention may contain 0.1%-95% of the pharmaceutical combination or a salt thereof of this invention, preferably 1 %-70%. In any event, the composition or formulation to be administered will contain a quantity of the pharmaceutical combination or a salt thereof according to the invention in an amount effective to treat the disease/condition of the subject being treated.
All documents cited in this application, including patents and patent applications, are hereby incorporated by reference. The examples presented below are intended to illustrate particular embodiments of the invention and are not intended to limit the invention, including the claims, in any manner.
Examples
The following abbreviations, when used in this application, have the following meanings. NMR nuclear magnetic resonance
H hydrogen s singlet d doublet t triplet m multiplet bm . broad multiplet
MS mass spectra
LCMS liquid chromatography mass spectrometry
APCI+ atmospheric pressure chemical ionization (positive mode) HPLC high pressure liquid chromatography
SEM standard error measurement
The pharmaceutical combinations of this invention generally will be administered in a convenient formulation. The following formulation examples only are illustrative and are not intended to limit the scope of the present invention. In the formulations that follow, "active ingredient" means a combination of a first compound, cis-6- phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof and a second compound, a selective androgen receptor modulator. In a preferred embodiment, the first compound of the "active ingredient" used in the formulation is (-)-cis-6-phenyl-5-(4- (2-pyrrolidin-1 -yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, tartrate salt. Formulation 1 : Gelatin Capsules
Hard gelatin capsules are prepared using the following:
Ingredient Quantity (mg/capsule)
Active ingredient 0.25-100
Starch, NF 0-650
Starch flowable powder 0-50
Silicone fluid 350 centistokes 0-15
A tablet formulation is prepared using the ingredients below: Formulation 2: Tablets
Ingredient Quantity (mg/tablet)
Active ingredient 0.25-100
Cellulose, microcrystalline 200-650
Silicon dioxide, fumed 10-650
Stearate acid 5-15
The components are blended and compressed to form tablets. Alternatively, tablets each containing 0.25-100 mg of active ingredients are made up as follows:
Formulation 3: Tablets
Ingredient Quantity (mg/tablet)
Active ingredient 0.25-100
Starch 45
Cellulose, microcrystalline 35
Polyvinylpyrrolidone (as 10% solution in water) 4
Sodium carboxymethyl cellulose 4.5
Magnesium stearate 0.5
Talc 1 The active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders which are then passed through a No. 14 mesh U.S. sieve. The granules so produced are dried at 50° - 60°C and passed through a No. 18 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 60 U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets.
Suspensions each containing 0.25-100 mg of active ingredient per 5 ml dose are made as follows: Formulation 4: Suspensions
Ingredient Quantity (mg/5 ml)
Active ingredient 0.25-100 mg
Sodium carboxymethyl cellulose 50 mg
Syrup 1.25 mg
Benzoic acid solution 0.1O mL
Flavor q.v.
Color q.v.
Purified Water to 5 mL
The active ingredient is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form smooth paste. The benzoic acid solution, flavor, and color are diluted with some of the water and added, with stirring. Sufficient water is then added to produce the required volume.
An aerosol solution is prepared containing the following ingredients: Formulation 5: Aerosol
Ingredient Quantity (% by weight)
Active ingredient 0.25
Ethanol 25.75
Propellant 22 (Chlorodifluoromethane) 70.00 The active ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 3O0C, and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remaining propellant. The valve units are then fitted to the container.
Suppositories are prepared as follows: Formulation 6: Suppositories
Ingredient Quantity (mg/suppository)
Active ingredient 250
Saturated fatty acid glycerides 2,000 The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimal necessary heat. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool. An intravenous formulation is prepared as follows: Formulation 7: Intravenous Solution
Ingredient Quantity
Active ingredient dissolved in ethanol 1% 20 mg
Intralipid™ emulsion 1 ,00O mL
The solution of the above ingredients is intravenously administered to a patient at a rate of about 1 ml. per minute.
Soft gelatin capsules are prepared using the following: Formulation 8: Soft Gelatin Capsule with Oil Formulation
Ingredient Quantity (mg/capsule)
Active ingredient 10-500
Olive Oil or Miglyol® Oil 500-1000
Preparation 1 Preparation of (+V2-ethyl-piperidine by resolution of 2-ethyl-piperidine
(R)-(I )-mandelic acid (40 g, 265 mmol.) and 2-ethyl-piperidine (30 g, 265 mmol.) were dissolved in methanol (10OmL). The mixture was warmed gently to ensure all material was in solution and then it was cooled to 0 °C. Diethyl ether (230 mL) was added slowly to the cooled solution and it was allowed to sit for 24 hours at 00C. The resulting white crystals were isolated and dried under high vacuum. The resulting salt was dissolved in warm methanol. Diethyl ether was added and the resulting solution was cooled to 0°C to afford the desired crystalline product. The isolated mandelic acid salt of (+)-2-ethyl-piperidine was dissolved in cooled H2O and solid potassium hydroxide was added to bring the pH of the solution to 14. The (+)-2-ethyl-piperidine (15.88 g) was extracted with diethyl ether (3x), dried (MgSO4), filtered, and concentrated to a clear oil. [α]589+4.88o (0.413g/mL, CHCI3). 1H NMR (CDCI3)δ:0.7 (m, 3H), 0.9 (m, 1H), 1.2 (bm, 3H), 1.3 (bm, 1 H), 1.4 (bm, 1 H), 1.6 (m, 1 H), 2.0 (bm, 1 H), 2.2 (bm, 1 H), 2.4 (bm, 1 H), 2.9 (bm, 1H).
Example 1 4-(2-(S)-Ethyl-piperidin-1-vO-2-trifluoromethyl-benzonitrile
(+)-2-Ethyl-piperidine (3.0 g, 26.5 mmol., Preparation 1) and 4-fluoro-2-trifluoromethyl-benzonitrile (2.0 g, 10.6 mmol.) were heated neat at 65°C overnight. The reaction mixture was cooled and partitioned between diethyl ether and 1 N HCI. The organic layers were combined, dried (MgSO4), filtered, and evaporated to dryness. The resulting residue (0.5 g, 2.64 mmol.) was a mixture of desired product and starting 4-fluoro-2-trifluoromethyl-benzonitrile. This mixture was treated with ethane-1 ,2-diamine (0.64 g, 10.58 mmol.) and heated at 80°C for 3 days. The reaction mixture was cooled and partitioned between diethyl ether and 0.5 N HCI. The organic layer was washed with 0.5N HCI (5x), dried (MgSO4), filtered, and evaporated to dryness. The resulting yellow oil was purified via Biotage™ Flash 40 (Biotage Inc.,
Charlottesville, VA, USA) chromatography using 10% ethyl acetate/hexanes as the eluant to afford the desired title product (11.8 g) as a yellow oil. MS (LCMS+) CaIc: 282.3, Found: 283.4 (M+1). 1H NMR (CDCI3)δ: 0.9 (t, 3H), 1.6 (m, 8H), 3.1 (m, 1H), 3.6 (bm, 1 H), 3.9 (bm, 1H), 6.9 (bm, 1H), 7.0 (s, 1H), 7.5 (m, 1H). Example 2
4-(2-(S)-Ethyl-piperidin-1-vπ-2,6-difluoro-benzonitrile A procedure analogous to the procedure described in Example 1 was followed by reacting 2,4,6- trifluoro-benzonitrile with (+)-2-ethyl-piperidine at 1050C to afford the crude product. The crude residue was purified by Chromatotron® centrifugal thin-layer chromatograph (2000μ, Harrison Research, Inc., Palo Alto, CA, USA) using 2% ethyl acetate/hexanes as the eluant to yield the title compound. MS (APCI+) CaIc: 250.2, Found: 251.2 (M+1). 1H NMR (CDCI3)δ: 0.9 (m, 3H), 1.7 (bm, 8H), 3.0 (m, 1 H), 3.5 (m, 1H), 3.8 (m, 1 H), 6.3 (m, 2H).
Example 3
2-Chloro-4-(2-(Sy-ethyl-piperidin-1-vD-benzonitrile A procedure analogous to the procedure described in Example 1 was followed by reacting 2- chloro-4-fluoro-benzonitrile with (+)-2-ethyl-piperidine at 1050C to afford the crude product. The crude residue was purified by Chromatotron® (2000μ) using 2% ethyl acetate/hexanes as the eluant to yield the title compound. MS (APCI+) CaIc: 248.8, Found: 249.2 (M+1). 1H NMR (CDCI3)δ: 0.9 (m, 3H), 1.7 (bm, 8H), 3.0 (m, 1H), 3.6 (m, 1H), 3.8 (m, 1H), 6.7 (m, 1H), 6.8 (m, 1 H), 7.4 (m, 1 H).
Example 4 4-(2-(S)-Ethyl-piperidin-1-vO-phthalonitrile
A procedure analogous to the procedure described in Example 1 was followed by reacting A- fluoro-phthalonitrile with (+)-2-ethyl-piperidine at 105°C to afford the crude product. The crude residue was purified by Chromatotron® (2000μ) using 2% ethyl acetate/hexanes as the eluant to yield the title compound. MS (APCI+) CaIc: 239.3, Found: 240.2 (M+1). 1H NMR (CDCI3)δ: 0.9 (m, 3H), 1.7 (bm, 8H), 3.0 (m, 1 H), 3.6 (m, 1H), 3.9 (m, 1H), 6.9 (m, 1 H), 7.0 (m, 1 H), 7.5 (m, 1H).
Example 5 4-(2-Ethyl-piperidin-1-vO-phthalonitrile
A procedure analogous to the procedure described in Example 1 was followed by reacting A- fluoro-phthalonitrile with 2-ethyl-piperidine at 750C to afford the crude product. The crude residue was purified by Chromatotron® (2000μ) using 2% ethyl acetate/hexanes as the eluant to yield the title compound. MS (APCI+) CaIc: 239.3, Found: 240.3 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 3H), 1.7 (bm, 8H), 3.1 (m, 1H), 3.6 (m, 1H), 3.9 (m, 1H), 6.9 (m, 1 H), 7.1 (m, 1 H), 7.5 (m, 1H).
Example 6 4-(2-Ethyl-piperidin-1-vO-2-trifluoromethyl-benzonitrile
The general procedure described in Example 1 was followed by reacting 4-fluoro-2-trifluoromethyl- benzonitrile with 2-ethyl-piperidine at 70°C to yield the desired crude product. The reaction mixture was cooled and partitioned between dichloromethane and 2M HCI. The organic layer was dried (MgSO4), filtered, and concentrated to dryness. The residue was purified by preparative thin-layer chromatography (TLC) using 30% ethyl acetate/hexanes as the eluant to give the title compound. MS (APCI+) CaIc: 282.3, Found: 283.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 3H), 1.7 (bm, 8H)1 3.0 (m, 1 H), 3.7 (m, 1 H), 3.9 (m, 1 H), 6.9 (m, 1 H), 7.1 (m, 1H), 7.6 (m, 1H).
Example 7
4-((RVsec-Butyl-methyl-amino)-2-trifluoromethyl-benzonitrile Step A: Preparation of 4-(R)-sec-Butylamino-2-trifluoromethyl-benzonitrile
Ethyl-diisopropyl-amine (7.1 g, 109.38 mmol.), (-)-sec-butylamine (6.0 g, 82.03 mmol.), and 4- fluoro-2-trifluoromethyl-benzonitrile (10.3 g, 54.69 mmol.) were combined and heated overnight at 600C. The reaction mixture was cooled and partitioned between diethyl ether and 1N HCI. The organic layer was dried (MgSO4), filtered, and concentrated to dryness. The resulting yellow liquid formed white crystals upon standing. The crystalline material was isolated by filtration and found to be pure desired product. The mother liquor was heated with hexanes and allowed to cool to ambient temperature for recrystallization. A total of 7.39 g of white crystalline product was obtained. 1H NMR (CDCI3) δ: 1.0 (t, 3H), 1.2 (d, 3H), 1.6 (m, 2H), 3.5 (m, 1H), 4.4 (bm, 1 H), 6.6 (m, 1 H), 6.8 (s, 1 H), 7.5 (m, 1H).
Step B: Preparation of 4-((R)-sec-Butyl-methyl-amino)-2-trifluoromethyl-benzonitrile
4-(R)-sec-butylamino-2-trifluoromethyl-benzonitrile (2.6 g, 10.73 mmol.) dissolved in tetrahydrofuran (THF) (6OmL) and the resulting solution was added to a flask containing a 60% dispersion of sodium hydride in mineral oil (0.515 g, 21.47 mmol.) and the mixture was stirred for 10 minutes at ambient temperature, lodomethane (3.05 g, 21.47 mmol.) was then added to the reaction mixture and stirring was continued at ambient temperature overnight. The reaction mixture was cooled to 00C, quenched with H2O, and extracted with diethyl ether. The organic layers were combined, washed with brine, dried (MgSO4), filtered, and concentrated to dryness. The crude material was purified via Biotage™ Flash 40 chromatography using 5% - 10% ethyl acetate/hexanes as the gradient eluant to obtain the title compound (2.5 g) as a light yellow oil. MS (LCMS+) CaIc: 256.3, Found: 257.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (t, 3H), 1.2 (m, 3H)1 1.6 (m, 2H), 2.8 (s, 3H), 3.9 (m, 1 H), 6.8 (m, 1H), 7.0 (m, 1 H), 7.5 (m, 1H).
Example 8
4-((R)-sec-Butyl-ethyl-amino)-2-chloro-benzonitrile
Step A: Preparation of 4-(R)-sec-Butylamino-2-chloro-benzonitrile
Following the general procedure in Example 7, Step A, 2-chloro-4-fluoro-benzonitrile was reacted with R-(-)-sec-butylamine to give the desired crude product. The crude material was purified by silica gel chromatography using 2% ethyl acetate/hexanes as the eluant to yield the desired pure product.
Step B: Preparation of 4-((R)-sec-Butyl-ethyl-amino)-2-chloro-benzonitrile
A 35% dispersion of potassium hydride in mineral oil (514 mg, 4.5 mmol.) was washed with hexanes (2x) under an inert atmosphere. 4-(R)-sec-butylamino-2-chloro-benzonitrile (308 mg, 1.5 mmol.) was dissolved in THF (5 mL) and added to the flask containing the potassium hydride under an inert atmosphere. The reaction mixture was stirred at ambient temperature and treated with iodoethane (0.24 mL, 3.0 mmol.). The reaction mixture was quenched slowly with H2O and extracted with diethyl ether. The organic layer was dried (MgSCvt), filtered, and evaporated to dryness. The crude product was purified by silica gel chromatography using 2% ethyl acetate/hexanes as the eluant to afford the desired compound. This material was further purified by Shimadzu® preparative HPLC (Shimadzu Scientific Instruments,
Columbia, MD, USA, a subsidiary of Shimadzu Corporation, Kyoto, Japan) using a Waters Symmetry® C8, 5μm, 19 mm x 50 mm column (Waters Corporation, Milford, MA, USA). The gradient eluant used was 15% to 0% acetonitrile, 0.1% formic acid in water to give the title compound. MS (APCI+) CaIc: 236.8, Found: 237.2 (M+1). 1H NMR (CDCI3) δ: 0.8 (m, 3H), 1.2 (m, 6H), 1.6 (m, 1H), 1.7 (m, 1H), 3.3 (m, 2H), 3.8 (m, 1H), 6.7 (m, 1H), 6.8 (m, 1H), 7.4 (m, 1H).
The compounds of Examples 9, 10, 12, 13, 14 and 27 were prepared by following the general procedure of Example 8, Step B, but substituting appropriate benzonitrile derivative and alkyl iodide. The compounds of Examples 15, 16, 17, 18, 19, 20 and 22-23 were prepared by following the general procedure of Example 8, Step A, but substituting the appropriate benzonitrile derivative and appropriate amine. Examples 11, 21 and 24-26 were prepared by following the general procedure of Example 8, Steps A and B, but substituting the appropriate benzonitrile derivative, amine and alkyl iodide.
Example 9 4-((R)-sec-Butyl-methyl-amino)-2-chloro-benzonitrile
4-(R)-sec-butylamino-2-chloro-benzonitrile was reacted with iodomethane to yield the title compound. MS (APCI+) CaIc: 222.7, Found: 223.2 (M+1). 1H NMR (CDCI3) δ: 0.8 (t, 3H), 1.1 (m, 3H), 1.6 (m, 2H), 2.7 (s, 3H), 3.8 (m, 1H), 6.6 (m, 1H), 6.7 (m, 1H), 7.4 (m, 1H).
Example 10
4-((R)-sec-Butyl-propyl-amino)-2-chloro-benzonitrile
4-(R)-sec-butylamino-2-chloro-benzonitrile was reacted with iodopropane to afford the title compound. MS (APCI+) CaIc: 250.8, Found: 251.2 (M+1). 1H NMR (CDCI3) δ: 0.8 (m, 6H), 1.2 (m, 3H), 1.6 (m, 4H), 3.1 (m, 2H), 3.8 (m, 1H), 6.5 (m, 1H), 6.7 (s, 1H), 7.4 (m, 1H).
Example 11
4-((R)-sec-Butyl-propyl-amino)-phthalonitrile Step A: Preparation of 4-(R)-sec-Butylamino-phthalonitrile 4-Fluoro-phthalonitrile was reacted with (-)-sec-butylamine to afford the desired compound.
Step B: Preparation of 4-((R)-sec-Butyl-propyl-amino)-phthalonitrile
4-(R)-sec-butylamino-phthalonitrile was reacted with iodopropane to afford the title compound. MS (APCI+) CaIc: 241.3, Found: 242.2 (M+1). 1H NMR (CDCI3) δ: 0.8 (t, 3H), 0.9 (t, 3H), 1.2 (d, 3H), 1.6 (bm, 4H), 3.1 (m, 2H), 3.8 (m, 1 H), 6.8 (m, 1 H), 6.9 (s, 1 H), 7.5 (m, 1 H). Exampie 12 4-((RVsec-Butyl-ethyl-aminoV2-trifluoromethyl-benzonitrile
4-(R)-sec-butylamino-2-trifluoromethyl-benzonitrile was reacted with iodoethane to afford the title compound. MS (APCI+) CaIc: 270.3, Found: 271.3 (M+1). 1H NMR (CDCI3) δ: 0.9 (t, 3H), 1.2 (m, 6H), 1.6 (m, 2H), 3.3 (m, 2H), 3.9 (m, 1 H), 6.8 (m, 1H), 6.9 (s, 1H)1 7.5 (m, 1H).
Example 13
4-((R)-sec-Butyl-methyl-aminoVphthalonitrile 4-(R)-sec-butylamino-phthalonitrile was reacted with iodomethane to afford the title compound.
MS (APCI+) CaIc: 213.2, Found: 214.2 (M+1). 1H NMR (CDCI3) δ: 0.82 (t, 3H), 1.20 (t, 3H), 1.60 (m, 1H), 3.83 (m, 1 H), 6.88 (dd, 1 H), 6.98 (d, 1 H), 7.52 (d, 1 H).
Example 14 4-(R)-sec-Butyl-ethyl-amino)-phthalonitrile
4-(R)-sec-butylamino-phthalonitrile was reacted with iodoethane to afford the title compound. MS (APCI+) CaIc: 227.3, Found 228.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (t, 3H), 1.2 (bm, 6H), 1.6 (bm, 2H), 3.3 (m, 2H), 3.8 (m, 1 H), 6.9 (m, 1H), 7.0 (s, 1 H), 7.5 (m, 1H).
Example 15
4-Dipropylamino-2-trifluoromethyl-benzonitrile
4-Fluoro-2-trifluoromethyl-benzonitrile was reacted with dipropylamine at 65°C to afford the title compound. MS (APCI+) CaIc: 270.3, Found: 271.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 6H), 1.6 (m, 4H)7 3.3
(m, 4H), 6.7 (m, 1H), 6.8 (m, 1H), 7.5 (m, 1 H).
Example 16
4-(Ethyl-isopropyl-amino)-phthalonitrile
4-Fluoro-phthalonitrile was reacted with ethyl-isopropyl-amine at 750C to afford the title compound. MS
(APCI+) CaIc: 213.3, Found: 214.2 (M+1). 1H NMR (CDCI3) δ: 0.87 (t, 3H), 1.6 (m, 6H), 3.1 (m, 1H), 3.6 (m, 1H), 3.92 (m, 1H), 6.98 (m, 1H), 7.02 (m,1 H), 7.52 (m, 1 H).
Example 17 4-(sec-Butyl-methyl-amino)-2-trifluoromethyl-benzonitrile
4-Fluoro-2-trifluoromethyl-benzonitrile was reacted with sec-butyl-methyl-amine at 700C to give the desired product. The crude product was purified via Chromatotron® (2000μ) using 30% ethyl acetate/hexanes as the eluant. MS (APCI+) CaIc: 256.3, Found: 257.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 3H), 1.2 (m, 3H), 1.6 (m, 2H), 2.8 (s, 1H), 3.9 (m, 1 H), 6.8 (m, 1 H), 7.0 (m, 1H), 7.6 (m, 1H). Example 18 4-(sec-Butyl-propyl-aminoVphthalonitrile
4-Fluoro-phthalonitrile was heated with sec-butyl-propyl-amine at 1050C to afford the title compound. MS (APCI+) CaIc: 241.3, Found: 242.3 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 3H), 1.0 (m, 3H), 1.2 (m, 3H), 1.6 (m, 4H), 3.1 (m, 2H), 3.8 (m, 1 H), 6.8 (m, 1H), 6.9 (m, 1 H), 7.5 (m, 1 H).
Example 19
4-Dipropylamino-phthalonitrile 4-Fluoro-phthalonitrile was heated with di-n-propyl amine at 1050C to afford the title compound.
MS (APCI+) CaIc: 227.3, Found: 228.2 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 6H), 1.6 (m, 4H), 3.3 (m, 4H), 6.7 (m, 1 H), 6.8 (m, 1 H), 7.5 (m, 1 H).
Example 20 4-Diethylamino-2-trifluoromethyl-benzonitrile
4-Fluoπ>2-trifluoromethyl-benzonitrile was reacted with diethyl amine at 65°C to afford the title compound. MS (APCI+) CaIc: 242.2, Found: 243.2 (M+1). 1H NMR (CDCI3) δ: 1.2 (m, 6H), 3.4 (m, 4H), 6.7 (m, 1H), 6.9 (m, 1H), 7.5 (m, 1 H).
Example 21
4-(sec-Butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile Step A: Preparation of 4-sec-butylamino-2-trifluoromethyl-benzonitrile
Fluoro-2-trifluoromethyl-benzonitrile was reacted with sec-butylamine to afford 4-sec-butylamino-2- trifluoromethyl-benzonitrile. Step B: Preparation of 4-(sec-butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile
4-sec-butylamino-2-trifluoromethyl-benzonitrile was reacted with iodoethane to yield the crude compound. The crude material was purified by Chromatotron® (4000μ) using 15% ethyl acetate/hexanes as the eluant to afford the title compound. MS (APCI+) CaIc: 270.3, Found: 271.4 (M+1). 1H NMR (CDCI3) δ: 0.9 (t, 3H), 1.2 (m, 6H), 1.6 (m, 2H), 3.3 (m, 2H), 3.9 (m, 1H), 6.8 (m, 1 H), 7.0 (s, 1H), 7.5 (m, 1H).
Example 22 2-Chloro-4-(ethyl-propyl-amino)-benzonitrile
2-Chloro-4-fluoro-benzonitrile was reacted with ethyl-propyl-amine at 900C to afford the title compound. MS (APCI+) CaIc: 222.7, Found: 223.2 (M+1). 1H NMR (CDCL3) δ: 0.9 (t, 3H), 1.1 (t, 3H), 1.6 (m, 2H), 3.2 (m, 2H), 3.4 (m, 2H), 6.5 (m, 1 H), 6.6 (s, 1 H), 7.4 (m, 1 H). Example 23 2-Chloro-4-(isopropyl-methyl-amino)-benzonitrile
2-Chloro-4-fluoro-benzonitrile was reacted with isopropyl-methyl-amine at 1050C to afford the title compound. MS (APCI+) CaIc: 208.7, Found: 209.1 (M+1). 1HNMR (CDCL3) δ: 1.2 (m, 6H), 2.8 (s, 3H), 4.1 (m, 1 H), 6.6 (m, 1 H), 7.4 (m, 1 H).
Example 24
4-Fd-(R). 2-Dimethyl-propyπ-methyl-aminol-2-trifluoromethyl-benzonitrile Step A: Preparation of 4-(1-(R). 2-dimethyl-propylamino)-2-trifluoromethyl-benzonitrile 4-Fluoro-2-trifluoromethyl-benzonitrile was reacted with 1-(R), 2-dimethyl-propylamine at 900C to afford the desired product.
Step B: Preparation of 4-Fd-(R), 2-dimethyl-propyl)-methyl-aminol-2-trifluoromethyl-benzonitrile
4-(1 ,2-dimethyl-propy!amino)-2-trifluoromethyl-benzonitrile was reacted with iodomethane to afford the title compound. MS (APCI+) CaIc: 270.3, Found 271.2 (M+1). 1H NMR (CDCI3) δ; 0.8 (d, 3H), 1.0 (d, 3H), 1.2 (d, 3H)1 1.8 (bm, 1 H), 2.8 (s, 3H), 3.6 (m, 1 H), 6.8 (m, 1H), 6.9 (m, 1 H), 7.5 (m, 1H).
Example 25
2-Chloro-4-F(1-(R), 2-dimethyl-propyl)-methyl-amino1-benzonitrile Step A: Preparation of 2-chloro-4-d-(R), 2-dimethyl-propylamino)-benzonitrile
2-Chloro-4-fluoro-benzonitrile was reacted with 1-(R), 2-dimethyl-propylamine at 900C to yield the desired product. Step B: Preparation of 2-chloro-4-F(1-(R). 2-dimethyl-propyl)-methyl-amino1-benzonitrile
2-Chloro-4-(1-(R), 2-dimethyl-propylamino)-benzonitrile was reacted with iodomethane to afford the title compound. MS (APCI+) CaIc: 236.8, Found: 237.2 (M+1). 1H NMR (CDCI3) δ: 0.8 (d, 3H), 1.0 (d, 3H), 1.2 (d, 3H), 1.8 (bm, 1H), 2.7 (s, 3H), 3.5 (m, 1H), 6.6 (m, 1H), 6.7 (s, 1H), 7.4 (m, 1H).
Example 26
4-(S)-sec-Butyl-methyl-amino)-2-trifluoromethyl-benzonitrile Step A: Preparation of 4-(S)-sec-butylamino-2-trifluoromethyl-benzonitrile
4-Fluoro-2-trifluoromethyl-benzonitrile was reacted with S-(+)-sec-butylamine to afford the desired product. Step B: Preparation of 4-((S)-sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile
4-(S)-sec-butylamino-2-trifluoromethyl-benzonitrile was reacted with iodomethane to afford the title compound. MS (APCI+) CaIc: 256.3, Found: 257.3 (M+1). 1H NMR (CDCI3) δ: 0.9 (t, 3H), 1.2 (d, 2H), 1.6 (m, 2H), 2.8 (s, 3H), 3.9 (m, 1 H), 6.8 (m, 1 H), 7.0 (m, 1H), 7.6 (m, 1H).
Example 27 4-((S)-sec-Butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile 4-(S)-sec-butylamino-2-trifluoromθthyl-benzonitrile was reacted with iodoethane to afford the title compound. MS (APCI+) CaIc: 270.3, Found: 271.3 (M+1). 1H NMR (CDCI3) δ: 0.9 (m, 3H), 1.2 (m, 6H), 3.3 (m, 2H), 3.9 (m, 1 H), 6.8 (m, 1H), 6.9 (s, 1 H), 7.6 (m, 1H).
Example 28 4-(2-Methyl-pyrrolidin-1-vO-2-trifluoromethyl-benzonitrile
4- fluoro-2-trifluoromethyl-benzonitrile (0.095 g, 0.5 mmol.) and 2-methyl-pyrrolidine (0.106 g, 1.25 mmol.) were heated neat at 6O0C for 3 days. The reaction mixture was cooled and partitioned between dichloromethane and 2M HCI, dried (MgSO4), filtered, and evaporated to dryness. The crude material was purified by preparative TLC using 30% ethyl acetate/hexanes as the eluant to afford the title compound. MS (APCI+) CaIc: 254.3, Found: 255.2 (M+1). 1H NMR (CDCI3) δ: 1.2 (m, 3H), 1.8 (m, 1H), 2.1 (m, 3H), 3.3 (m, 1H), 3.5 (m, 1 H), 4.0 (m, 1 H), 6.6 (m, 1 H), 6.8 (m, 1 H), 7.6 (m, 1 H).
Example 29 4-(Cvclopentyl-methyl-amino)-2-trifluoromethyl-benzonitrile
Following the general procedure in Example 28, 4-fluoro-2-trifluoromethyl-benzonitrile was reacted with cyclopentyl-methyl-amine at 55°C overnight to give the desired product. The crude material was purified by preparative TLC using 20% ethyl acetate/hexanes as the eluant to afford the title compound. MS (APCI+) CaIc: 268.3, Found: 269.2 (M+1). 1H NMR (CDCI3) δ: 1.7 (bm, 8H), 2.9 (m, 3H), 4.2 (bm, 1H), 6.8 (m, 1H), 6.9 (m, 1H), 7.5 (m, 1H).
Examples 30-53 Table 1 , below, provides Examples 30-53. Examples 30 through 53 can be prepared by methods analogous to the methods employed for the preparation of Examples 1-29. The compounds of Examples 30-53 can be purified and characterized according to methods analogous to the methods used for Examples 1-29.
Table 1
Figure imgf000027_0001
Figure imgf000028_0001
All salts of the Formula I compound are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate. The salts are recovered either by filtration, by precipitation with a non-solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate. The compounds can be obtained in crystalline form by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
In addition, when the Formula I compound used in the compositions and methods of this invention forms hydrates or solvates they are also within the scope of the invention.
The Formula I compound used in the compositions and methods of this invention, and the salts thereof are all adapted to therapeutic use as agents that mediate androgen receptors in mammals, particularly humans. By virtue of this activity, these agents are useful for treating conditions that present with low bone mass and improve frailty and other disease/conditions detailed above.
The utility of the Formula I compound of the invention and the salts thereof as medical agents in the treatment of the above described disease/conditions in mammals (e.g., humans, male or female) is demonstrated by the activity of the compound of this invention in conventional assays and the in vitro and in vivo assays described below. The in vitro and in vivo assays (with appropriate modifications within the skill in the art) may be used to determine the activity of analogous agents as well as the compounds of this invention. Such assays also provide a means whereby the activities of the Formula I compound of this invention, and the salts thereof can be compared with the activities of other known compounds. The results of these comparisons are useful for determining dosage levels in mammals, including humans, for the treatment of such diseases.
The following protocols can be varied when appropriate by those skilled in the art. Human Androgen Receptor Binding Analysis
The following is a brief description of the assay that determines the affinity of a compound for the recombinant human androgen receptor (hAR). Competitive binding analysis is performed on baculovirus/Sf9 generated hAR extracts in the presence or absence of differing concentrations of drug and a fixed concentration of 3H-dihydrotestosterone (3H-DHT) as tracer. This binding assay method is a modification of a protocol previously described (Chang, et. al. J. Steroid Biochem. 20(1):11-17 1984). Briefly, progressively decreasing concentrations of compounds are incubated in the presence of hAR extract (Chang et al. P.N.A.S. Vol. 89, pp. 5546-5950, 1992), hydroxylapatite and 1 nM 3H-DHT for one hour at 4°C. Subsequently, the binding reactions are washed three times to completely remove excess unbound 3H-DHT. hAR bound 3H-DHT levels are determined in the presence of compounds (= competitive binding) and compared to levels bound when no competitor is present (= maximum binding). Compound binding affinity to the hAR is expressed as the concentration of compound at which one half of the maximum binding is inhibited (the IC50).
Table 2, below, provides data obtained for compounds of the invention using the human androgen receptor binding analysis assay described hereinabove.
Table 2
Figure imgf000029_0001
Figure imgf000030_0001
Effect of Selective Androgen Receptor Modulator on Body Weight, Body Composition and Bone Density in the Aged Intact and Ovariectomized Female Rat
The purpose of this study is to test the effects of test compound in aged intact or ovariectomized (OVX) female rat model.
Study Protocol Sprague-Dawley female rats were sham-operated or OVX at 18 months of age, while a group of rats was necropsied at day 0 to serve as baseline controls. One day post-surgery, the rats were treated with either vehicle or SARM test compound, SERM test compound or a combination of SARM and SERM test compound for 59 days. The vehicle or SARM test compound was administered twice a week (Tuesday and Friday) by subcutaneous injection (s.α), with the SARM test compound being administered at an average dose of 10 milligrams per kilogram of body weight per day (10 mg/kg/day). The SERM test compound was administered daily at a dose of 0.1 milligrams per kilogram of body weight by oral gavage (p.o.).
All rats were given s.c. injection of 10 mg/kg of calcein (Sigma, St.Louis, MO) for fluorescent bone label 2 and 12 days before necropsy. On the day of necropsy, all rats under ketamine/xylazine anesthesia were weighed and underwent dual-energy X-ray absorptiometry (DXA, QDR-4500/W, Hologic Inc., Waltham, MA) equipped with Rat Whole Body Scan software for lean and fat body mass determination. The rats were necropsied, then autopsied and blood was obtained by cardiac puncture. The distal femoral metaphysis and femoral shafts from each rat were analyzed by peripheral quantitative computerized tomography (pQCT), and volumetric total, trabecular and cortical bone mineral content and density were determined. Peripheral Quantitative Computerized Tomography (pQCT) Analysis: Excised femurs were scanned by a pQCT X-ray machine (Stratec XCT Research M, Norland Medical Systems, Fort Atkinson, Wl.) with software version 5.40.' A 1 millimeter (mm) thick cross section of the femur metaphysis was taken at 5.0 mm (proximal femoral metaphysis, a primary cancellous bone site) and 13 mm (femoral shafts, a cortical bone site) proximal from the distal end with a voxel size of 0.10 mm. Cortical bone was defined and analyzed using contour mode 2 and cortical mode 4. An outer threshold setting of 340 mg/cm3 was used to distinguish the cortical shell from soft tissue and an inner threshold of 529 mg/cm3 to distinguish cortical bone along the endocortical surface. Trabecular bone was determined using peel mode 4 with a threshold of 655 mg/cm3 to distinguish (sub)cortical from cancellous bone. An additional concentric peel of 1% of the defined cancellous bone was used to ensure that (sub)cortical bone was eliminated from the analysis. Volumetric content, density, and area were determined for both trabecular and cortical bone (Jamsa T. et al., Bone 23:155-161 , 1998; Ke, H.Z. et al., Journal of Bone and Mineral Research, 16:765-773, 2001). Using the above setting, it was determined that the ex vivo precision of volumetric content, density and area of total bone, trabecular, and cortical regions ranged from 0.99% to 3.49% with repositioning.
Vaginal histology: Vaginal tissue was fixed and embedded in paraffin. Five micron sections were cut and stained with Alcian Blue staining. Histology examination of vaginal luminal epithelial thickness and mucopolysaccharide (secreted cells) was performed.
The experimental groups for the protocol are as follows: Group I: Baseline controls Group II: Sham + Vehicle
Group III: OVX + Vehicle Group IV: OVX + SARM Test Compound at 10 mg/kg/day (in Vehicle)
Group V: OVX + SERM Test Compound at 0.1 mg/kg/day
Group Vl: OVX + SARM Test Compound at 10 mg/kg/day and SERM Test Compound at 0.1 mg/kg/day.
Study Results The preceding protocol was carried out using 4-(2-(S)-ethyl-piperidin-1-yl)-2-trifluoromethyl- benzonitrile, the compound of Example 1 , as the SARM test compound. The vehicle employed in experimental groups H-IV was 100% sesame oil. The rats in groups H-IV were dosed s.c. twice a week (on Tuesday and Friday) with 0.3 ml and 0.4 ml. This dosing provided an average daily dose of 10 mg/kg/day of the compound of Example 1 for experimental group IV. The SERM compound employed in experimental groups V and Vl is cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,- tetrahydronaphthalene-2-ol. The vehicle employed for the SERM test compound is one milliliter of 0.5% methylcellulose. The results are provided below in Table 3.
Figure imgf000032_0001
The values provided in Table 3 are the mean values ± the standard error measurement. In Table 3 and the experimental protocol the following abbreviations or symbols had the following definitions. DFM: distal femoral metaphysis; FS: femoral shafts; g: grams; mg/cm3: milligrams per cubic centimeter; mg/mm: milligrams per millimeter; a: p < 0.05 vs. Baseline; b: p < 0.05 vs. Sham; c: p < 0.05 vs. OVX; d: p < 0.05 vs. SARM compound; e: p < 0.05 vs. SERM compound; mg/kg/day: milligrams per kilogram of body weight per day; and ml: milliliter. Body weight and body composition: As shown in Table 3, OVX rats treated with 4-(2-(S)-ethyl- piperidin-1-yl)-2-trifluoromethyl-benzonitrile (SARM) had little effect on body weight, while cis-6-phenyl- 5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol (SERM) and the combination of SARM and SERM decreased body weight as compared with vehicle-treated OVX controls. Fat body mass decreased significantly in SARM alone and SERM alone groups as compared with vehicle treated OVX controls. Combination of SARM and SERM further decreased fat body mass as compared with either SARM alone or SERM alone groups. Lean body mass increased significantly in the SARM alone or in combination groups, while it did not differ significantly in the SERM group as compared with vehicle treated OVX controls. These data indicate that combination therapy with a SARM and a SERM significantly increased lean body mass and decreased fat body mass.
Distal femoral pQCT analysis: PQCT analysis of distal femoral metaphysis showed that total mineral density and trabecular mineral density of distal femoral metaphysis significantly decreased in vehicle- treated OVX group as compared to vehicle treated sham controls. Total mineral density and trabecular mineral density of distal femoral metaphysis increased significantly in cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl- ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphthalene-2-ol (SERM) treated OVX rats or in the SERM and SARM combination group as compared with vehicle treated OVX controls. These two parameters did not differ between SARM treated OVX rats compared with vehicle treated OVX controls.
Femoral shaft pQCT analysis: PQCT analysis of femoral shaft showed that total mineral content and cortical mineral content of distal femoral metaphysis significantly decreased in vehicle-treated OVX group as compared to vehicle treated sham controls. Total mineral content and cortical mineral content of femoral shafts increased significantly in SARM treated OVX rats or in the SERM and SARM combination group as compared with vehicle treated OVX controls. These two parameters did not differ significantly between SERM treated OVX rats compared with vehicle treated OVX controls.
Effects on vaginal histology: OVX caused dramatically thinning of vaginal luminal epithelial and decrease in mucopolysaccharide (secreted cells) staining. Treatment with cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl- ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphtha-lene-2-ol (SERM) partially prevented these changes induced by OVX, while treatment with 4-(2-(S)-ethyI-piperidin-1-yl)-2-trifluoromethyl-benzonitrile (SARM) partially prevented the thinning of vaginal luminal epithelium induced by OVX. However, combination of cis-6- phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphtha-lene-2-ol (SERM) and 4-(2-(S)- ethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile (SARM) completely prevented these changes induced by OVX. This data indicates that combination therapy with cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- phenyl)-5,6,7,8,-tetrahydronaphtha-lene-2-ol (SERM) and a SARM has beneficial effects in female sexual dysfunction since this treatment prevents against the dryness of vaginal tissue induced by estrogen deficiency.
Summary and Conclusion
These data reveal that combination of a SARM and a SERM significantly increased both trabecular and cortical bone mass, while SARM increased only the cortical bone and the SERM increased only the trabecular bone in rat model of osteoporosis, as compared with vehicle controls. Thus the combination treatment provides more benefits than mono-therapy. In addition, combination treatment provides further decrease in fat body mass compared with mono-therapy. Furthermore, combination of cis-
6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8,-tetrahydronaphtha-lene-2-ol (SERM) and 4-(2-(S)- ethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile (SARM) completely prevented the thinning of vaginal luminal epithelium and the decrease in mucopolysaccharide (secreted cells) staining induced by estrogen deficiency (OVX) in aged female rats, indicating that this treatment may have beneficial effects in female sexual dysfunction. Orchiectomized Immature Rat Assay
All animal studies were performed in accordance with the Guide for Care and Use of Laboratory Animals (National Research Council, 1996). Twenty-one day old male Sprague Dawley rats were acquired from Taconic, Inc, Germantown, NY. Rats were housed five per cage at standard vivarium conditions (72°C, 12-h light/dark cycle), with normal chow diet and tap water ad libitum. At 25 days of age, the rats underwent bilateral orchiectomy surgery (ORX) after sedation with 3-5% Isoflurane. Following the surgery, the animals were treated immediately by daily subcutaneous (s.c.) injection with vehicle (sesame oil), testosterone propionate (TP, 10 mg/kg) and test compounds. The dosage of test compound administered is in mg/kg. At the end of the 4 days of administrations, the animals were sacrificed in a carbon dioxide chamber, the ventral prostate (VP) and levator ani muscle complex (LA) were removed and weighed. The increase in the weights of VP or LA by TP was expressed as 100%, and the ORX control as baseline, 0 %; the response of the test compounds was calculated as the percentage increase compared to TP. Experimental results obtained for the compounds of Examples 1 and 7 dosed at 3, 10 and 30 mg/kg are provided in Table 4, below.
Table 4
Figure imgf000034_0001
Effect of Selective Androgen Receptor Modulator on Body Weight. Body Composition and Bone Density in the Aged Intact and Orchidectomized Male Rat
The purpose of this study is to test the effects of test compound in aged intaGt or orchidectomized (ORX) male rat model.
Study Protocol
Male SD rats at 11 months of age were sham-operated or ORX. One day post-surgery they were treated with test compound by subcutaneous injections (s.c.) at the average dose of 3 or 10 mg/kg per day for 8 weeks. The subcutaneous injections were given 2 times (Tuesday and Friday) per week with the first injection (Tuesday) at 9 or 30 mg/kg (in sesame oil, total 0.3 ml), and the second injection (Friday) at 12 or 40 mg/kg (in sesame oil, total 0.4 ml). All rats were injected subcutaneously with calcein at 10 mg/kg on 12 and 2 days before necropsy. The experimental groups are as follows:
Group I: Sham + Vehicle G Grroouupp I III:: Sham + Test Compound at 3 mg/kg/d
Group III: Sham + Test Compound at 10 mg/kg/d
Group IV: ORX + Vehicle
Group V: ORX + Test Compound at 3 mg/kg/d
Group Vl: ORX + Test Compound at 10 mg/kg/d
S Sttuuddyv F Results
The preceding protocol was carried out using the compound of Example 1 as the test compound.
Treatment with the compound of Example 1 according to the above protocol decreased fat body mass and increased lean body mass in both sham and ORX rats in a dose dependant manner, with the exception of Group Il which showed a slight increase in fat body mass and slight decrease in lean body mass compared to Group I. Treatment with the compound of Example 1 increased trabecular density in the distal femoral metaphysis (DFM) and total density in the femoral shaft (FS) in both sham and ORX rats.
Treatment with the compound of Example 1 increased levitor anni weight in both sham and ORX rats.
Treatment with the compound of Example 1 did not increase prostate weight in Group Il sham rats, although there was a slight increase in prostate weight of Group III compared to Group I. Treatment with the compound of Example 1 increased prostate weight in ORX rats to the level of sham controls in a dose dependent manner.
Fracture Healing Assays Assay For Effects On Fracture Healing After Systemic Administration
Fracture Technioue: Sprage-Dawley rats at 3 months of age are anesthetized with Ketamine. A 1 cm incision is made on the anteromedial aspect of the proximal part of the right tibia or femur. The following describes the tibial surgical technique. The incision is carried through to the bone, and a 1 mm hole is drilled 4 mm proximal to the distal aspect of the tibial tuberosity 2 mm medial to the anterior ridge. Intramedullary nailing is performed with a 0.8 mm stainless steel tube (maximum load 36.3 N, maximum stiffness 61.8 N/mm, tested under the same conditions as the bones). No reaming of the medullary canal is performed. A standardized closed fracture is produced 2 mm above the tibiofibular junction by three-point bending using specially designed adjustable forceps with blunt jaws. To minimize soft tissue damage, care is taken not to displace the fracture. The skin is closed with monofilament nylon sutures. The operation is performed under sterile conditions. Radiographs of all fractures are taken immediately after nailing, and rats with fractures outside the specified diaphyseal area or with displaced nails are excluded. The remaining animals are divided randomly into the following groups with 10 - 12 animals per each subgroup per time point for testing the fracture healing. The first group receives daily gavage of vehicle (water : 100% Ethanol = 95 : 5) at 1 ml/rat, while the others receive daily gavage from 0.01 to 100 mg/kg/day of the pharmaceutical composition to be tested (1 ml/rat) for 10, 20, 40 and 80 days.
At 10, 20, 40 and 80 days, 10 - 12 rats from each group are anesthetized with Ketamine and sacrificed by exsanguination. Both tibiofibular bones are removed by dissection and all soft tissue is stripped. Bones from 5 - 6 rats for each group are stored in 70% ethanol for histological analysis, and bones from another 5 - 6 rats for each group are stored in a buffered Ringer's solution (+4°C, pH 7.4) for radiographs and biomechanical testing which is performed.
Histological Analysis: The methods for histologic analysis of fractured bone have been previously published by Mosekilde and Bak (The Effects of Growth Hormone on Fracture Healing in Rats: A Histological Description. Bone, 14:19-27, 1993). Briefly, the fracture site is sawed 8 mm to each side of the fracture line, embedded undecalcified in methymethacrylate, and cut frontals sections on a Reichert-Jung Polycut microtome in 8 μm thick. Masson-Trichrome stained mid-frontal sections (including both tibia and fibula) are used for visualization of the cellullar and tissue response to fracture healing with and without treatment. Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site. The following measurements are performed: (1) fracture gap - measured as the shortest distance between the cortical bone ends in the fracture, (2) callus length and callus diameter, (3) total bone volume area of callus, (4) bony tissue per tissue area inside the callus area, (5) fibrous tissue in the callus, and (6) cartilage area in the callus.
Biomechanical Analysis: The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue lnt 45:292-297, 1989). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedure. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.
Assay for Effects on Fracture Healing After Local Administration
Fracture Technique: Female or male beagle dogs at approximately 2 years of age are used under anesthesia in the study. Transverse radial fractures are produced by slow continuous loading in three-point bending as described by Lenehan et al. (Lenehan, T. M.; Balligand, M.; Nunamaker, D. M.; Wood, F.E.: Effects of EHDP on Fracture Healing in Dogs. J Orthop Res 3:499-507; 1985). A wire is pulled through the fracture site to ensure complete anatomical disruption of the bone. Thereafter, local delivery of the test pharmaceutical composition to the fracture site is achieved by slow release of compound delivered by slow release pellets or by administration of the compounds in a suitable formulation such as a paste gel solution or suspension for 10, 15, or 20 weeks.
Histological Analysis: The methods for histologic analysis of fractured bone have been previously published by Peter et al. (Peter, C. P.; Cook, W.O.; Nunamaker, D. M.; Provost, M. T.; Seedor, J.G.; Rodan, G.A. Effects of alendronate on fracture healing and bone remodeling in dogs. J. Orthop. Res. 14:74-70, 1996) and Mosekilde and Bak (The Effects of Growth Hormone on Fracture Healing in Rats: A Histological Description. Bone, 14:19-27, 1993). Briefly, after sacrifice, the fracture site is sawed 3 cm to each side of the fracture line, embedded undecalcified in methymethacrylate, and cut on a Reichert-Jung Polycut microtome in 8 μm thick of frontal sections. Masson-Trichrome stained mid-frontal sections (including both tibia and fibula) are used for visualization of the cellullar and tissue response to fracture healing with and without treatment. Sirius red stained sections are used to demonstrate the characteristics of the callus structure and to differentiate between woven bone and lamellar bone at the fracture site. The following measurements are performed: (1) fracture gap - measured as the shortest distance between the cortical bone ends in the fracture, (2) callus length and callus diameter, (3) total bone volume area of callus, (4) bony tissue per tissue area inside the callus area, (5) fibrous tissue in the callus, (6) cartilage area in the callus.
Biomechanical Analysis: The methods for biomechanical analysis have been previously published by Bak and Andreassen (The Effects of Aging on Fracture Healing in Rats. Calcif Tissue lnt 45:292-297, 1989) and Peter et al. (Peter, C.P.; Cook, W.O.; Nunamaker, D.M.; Provost, M. T.; Seedor, J.G.; Rodan, G.A. Effects of Alendronate On Fracture Healing And Bone Remodeling In Dogs. J. Orthop. Res. 14:74-70, 1996). Briefly, radiographs of all fractures are taken prior to the biomechanical test. The mechanical properties of the healing fractures are analyzed by a destructive three- or four-point bending procedures. Maximum load, stiffness, energy at maximum load, deflection at maximum load, and maximum stress are determined.

Claims

CLAlMS
1. A pharmaceutical composition comprising: a. a first compound, said first compound being cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)- pheny!)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof; and b. a second compound, said second compound being a selective androgen receptor modulator, or a pharmaceutically acceptable salt thereof.
2. The pharmaceutical composition of claim 1 additionally comprising a pharmaceutical carrier or diluent.
3. The pharmaceutical composition of claim 1 wherein said first compound is (-)-cis-6-phenyl-5-(4-(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, or a pharmaceutically acceptable salt thereof.
4. The pharmaceutical composition of claim 3 wherein said first compound is (-)-cis-6-phenyl-5-(4-
(2-pyrrolidin-1-yl-ethoxy)-phenyl)-5,6,7,8-tetrahydronapthalene-2-ol, D-tartrate salt.
5. The pharmaceutical composition of claim 1 wherein said second compound is cyproterone, chlormadinone, flutamide, hydroxyflutamide, bicalutamide, nilutamide, or spironolactone, or a pharmaceutically acceptable salt thereof.
6. The pharmaceutical composition of claim 1 wherein said second compound is of the Formula I:
Figure imgf000038_0001
a prodrug thereof or a pharmaceutically acceptable salt of said compound or said prodrug, wherein:
R1 is hydrogen;
R2 is chloro, cyano or trifluoromethyl; or R1 and R2 are each fluoro;
R3 and R4 are each independently (CrC6)alkyl, (C3-C7)cycloalkyl or (C2-C6)alkenyl; or R3 and R4 taken together with the nitrogen to which they are attached is
Figure imgf000038_0002
n is 1 , 2 or 3; R5 is (CrC6)alkyl optionally substituted with hydroxy or (CrC6)alkoxy; and
R6 is hydrogen or (Ci-C6)alkyl optionally substituted with hydroxy or (CrC6)alkoxy.
7. The pharmaceutical composition of claim 1 wherein said second compound is: 4-(2-ethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-ethyl-piperidin-1-yl)-2,6-difluoro-benzonitrile;
2-chloro-4-(2-ethyl-piperidin-1-yl)-benzonitrile;
4-(2-ethyl-piperidin-1-yl)-phthalonitrile;
4-(sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile; 4-(sec-butyl-ethyl-amino)-2-chloro-benzonitrile;
4-(sec-butyl-methyl-amino)-2-ch!oro-benzonitrile;
4-(sec-butyl-propyl-amino)-2-chloro-benzonitriIe;
4-(sec-butyl-propyl-amino)-phthalonitrile;
4-(sec-butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile; 4-(sec-butyl-methyl-amino)-phthalonitrile;
4-(sec-butyl-ethyl-amino)-phthalonitrile;
4-dipropylamino-2-trifluoromethyl-benzonitrile;
4-(ethyl-isopropyl-amino)-phthalonitrile;
4-dipropylamino-phthalonitrile; 4-diethylamino-2-trifluoromethyl-benzonitrile;
2-chloro-4-(ethyl-propyl-amino)-benzonitrile;
2-chloro-4-(isopropyl-methyl-amino)-benzonitrile;
4-[(1 ,2-dimethyl-propyl)-methyl-amino]-2-trifluoromethyl-benzonitrile;
2-chloro-4-[(1 ,2-dimethyl-propyl)-methyl-amino]-benzonitrile; 4-(2-methyl-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(cyclopentyI-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(2-propyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-ethyl-4-hydroxymethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(sec-butyl-propyl-amino)-2-trifluoromethyl-benzonitrile; 4-(allyl-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(2-methyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-(ethoxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-(2-hydroxyethyl)-piperidin-1-yl)-2-trifluoromethyl-benzonitrile;
4-(2-(methoxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile; 4-(ethyl-pentyl-amino)-2-trifluoromethyI-benzonitrile;
4-(butyl-propyl-amino)-2-trifluoromethyl-benzonitrile;
4-(isopropyl-methyl-amino)-2-trifluoromethyl-benzonitrile;
4-(pentyl-propyl-amino)-2-trifluoromethyl-benzonitrile;
4-(2-hydroxymethyI-piperidin-1-yl)-2-trifluoromethyl-benzonitrile; 4-(butyl-ethyl-amino)-2-trifluoromethyl-benzonitrile; 4-(dibutyl-amino)-2-trifluoromethyI-ben2onitrile; 4-(2-(hydroxy-methyl)-pyrrolidin-1-yl)-2-trifluoromethyl-benzonitrile; 2-chloro-4-(isopropyl-propyl-amino)-benzonitrile; 2-chloro-4-(diethyl-amino)-benzonitrile; & 2-chloro-4-(ethyl-isopropyl-amino)-benzonitriIe;
2-chloro-4-(dipropyl-amino)-benzonitrile; 4-(isopropyl-methyl-amino)-phthalonitrile; 4-(2-methoxymethyl-pyrrolidin-1-yI)-phthalonitrile; 4-azepan-1-yl-phthalonitrile; or 0 2,6-difluoro-4-(dipropyl-amino)-benzonitrile; or a stereoisomer thereof or a pharmaceutically acceptable salt of said compound or stereoisomer.
8. The pharmaceutical composition of claim 1 wherein said second compound is 4-(2-(S)-ethyl-piperidin-1-yl)-2-trifluoromethyl-benzonitrile or 5 4-((R)-sec-butyl-methyl-amino)-2-trifluoromethyl-benzonitrile, or a pharmaceutically acceptable salt thereof.
9. A method of treating a condition that presents with low bone mass, treating a wasting disease, increasing muscle mass, increasing lean body mass, decreasing fat body mass, treating bone fracture and 0 muscle damage, treating prostate hypertrophy or treating female sexual dysfunction in a mammal, the method comprising administering to said mammal in need thereof a therapeutically effective amount of a pharmaceutical composition according to claim 1.
10. The method of claim 10 wherein the mammal has a condition that presents with low bone mass. 5
11. The method of claim 10 wherein the condition that presents with low bone mass is osteoporosis, a bone defect, childhood idiopathic bone loss, alveolar bone loss, mandibular bone loss, bone fracture, osteotomy, periodontitis or prosthetic ingrowth.
0 12. The method of claim 9 wherein concomitant bone fracture and muscle damage are treated.
13. The method of claim 9 wherein muscle mass is increased, lean body mass is increased or fat body mass is decreased.
5 14. The method of claim 13 wherein the mammal is a livestock mammal.
15. The method of claim 9 wherein female sexual dysfunction is treated and the therapeutically effective amount of the pharmaceutical composition is about 0.01 mg/kg/day to about 20 mg/kg/day.
PCT/IB2005/002592 2004-08-31 2005-08-19 Therapeutic combinations comprising a selective estrogen receptor modulator and a selective androgen receptor modulator WO2006024931A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60592504P 2004-08-31 2004-08-31
US60/605,925 2004-08-31

Publications (2)

Publication Number Publication Date
WO2006024931A2 true WO2006024931A2 (en) 2006-03-09
WO2006024931A3 WO2006024931A3 (en) 2006-05-26

Family

ID=35735140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2005/002592 WO2006024931A2 (en) 2004-08-31 2005-08-19 Therapeutic combinations comprising a selective estrogen receptor modulator and a selective androgen receptor modulator

Country Status (2)

Country Link
TW (1) TW200621231A (en)
WO (1) WO2006024931A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2447061C1 (en) * 2010-10-22 2012-04-10 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Ярославский государственный технический университет"(ЯГТУ) Novel 4-(azacycloalkyl)phthalonitriles and synthesis method thereof
US20150071857A1 (en) * 2012-03-07 2015-03-12 Ligand Pharmaceuticals, Inc. Steroid hormone and cholesterol pathways as one unified homeostatic system
EP3613418A1 (en) * 2014-01-17 2020-02-26 Ligand Pharmaceuticals, Inc. Methods and compositions for modulating hormone levels
WO2021098809A1 (en) * 2019-11-20 2021-05-27 南京明德新药研发有限公司 Bicyclic compound used as selective androgen receptor modulator

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999065486A1 (en) * 1998-06-16 1999-12-23 Pfizer Products, Inc. Therapeutic combinations of (selective) estrogen receptor modulators (serm) and growth hormone secretagogues (ghs) for treating musculoskeletal frailty
WO2003092588A2 (en) * 2002-04-30 2003-11-13 Merck & Co., Inc. 4-azasteroid derivatives as androgen receptor modulators

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999065486A1 (en) * 1998-06-16 1999-12-23 Pfizer Products, Inc. Therapeutic combinations of (selective) estrogen receptor modulators (serm) and growth hormone secretagogues (ghs) for treating musculoskeletal frailty
WO2003092588A2 (en) * 2002-04-30 2003-11-13 Merck & Co., Inc. 4-azasteroid derivatives as androgen receptor modulators

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2447061C1 (en) * 2010-10-22 2012-04-10 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Ярославский государственный технический университет"(ЯГТУ) Novel 4-(azacycloalkyl)phthalonitriles and synthesis method thereof
US20150071857A1 (en) * 2012-03-07 2015-03-12 Ligand Pharmaceuticals, Inc. Steroid hormone and cholesterol pathways as one unified homeostatic system
EP3613418A1 (en) * 2014-01-17 2020-02-26 Ligand Pharmaceuticals, Inc. Methods and compositions for modulating hormone levels
US10874638B2 (en) 2014-01-17 2020-12-29 Ligand Pharmaceuticals Incorporated Methods and compositions for modulating hormone levels
WO2021098809A1 (en) * 2019-11-20 2021-05-27 南京明德新药研发有限公司 Bicyclic compound used as selective androgen receptor modulator
CN114728904A (en) * 2019-11-20 2022-07-08 南京明德新药研发有限公司 Bicyclic compounds as selective androgen receptor modulators

Also Published As

Publication number Publication date
WO2006024931A3 (en) 2006-05-26
TW200621231A (en) 2006-07-01

Similar Documents

Publication Publication Date Title
US20080045504A1 (en) Benzonitrile Derivatives to Treat Musculoskeletal Frailty
CN102351732B (en) Nuclear receptor binding agents
JP4764719B2 (en) Halogenated selective androgen receptor modulator and method of use thereof
JP5763609B2 (en) 1- [2- (2,4-Dimethylphenylsulfanyl) -phenyl] piperazine as a compound with combined serotonin reuptake, 5-HT3 and 5-HT1A activity for the treatment of cognitive impairment
AP713A (en) (-) cis-6(s)-phenyl-5 (R) (4-(2-pyrrolidin-1-yl ethoxy)phenyl)-5,6,7,8,- tetrahydronaphthalen-2-ol-d- tartrate.
EP1285658A2 (en) Treatments for female sexual dysfunction
RU2001101487A (en) MEDICAL APPLICATION OF A SELECTIVE MODULATOR OF ESTROGEN RECEPTORS IN COMBINATION WITH PRECONDENTS OF SEXUAL STEROID HORMONES
CN1298409A (en) 20-keto-11&amp;beta, -arylsteroids and their derivatives having agonist or antagonist hormonal properties
JPH0680017B2 (en) Process for producing composition comprising anti-estrogen and carrier
KR100880564B1 (en) Method for the treatment of climacteric disorders in women during or after the menopause
JP2007533686A (en) Novel pharmaceutical composition for the treatment of sexual disorder II
JP5611991B2 (en) Use of estrogen receptor ligands in the manufacture of a medicament for ADT
CA2295640A1 (en) Aroylpiperazines for modulating sexual activity
JP2010522714A (en) Novel pharmaceutical composition
US10052317B2 (en) Method for treating or preventing hot flushes
WO2006024931A2 (en) Therapeutic combinations comprising a selective estrogen receptor modulator and a selective androgen receptor modulator
JP2013520445A (en) Estrogen receptor ligands and methods of use thereof
JP2017531012A (en) Method of treating urological disorders using SARM
JP2006306872A (en) Pharmaceutical composition comprising combination of selective estrogen receptor modifier and aromatase inhibitor and method
KR102024493B1 (en) Treatment of androgen deprivation therapy associated symptoms
JPWO2004035089A1 (en) Treatment for hormone-dependent cancer
MX2007002377A (en) Androgen modulators.
JP2004531559A (en) Tetrahydroquinoline derivatives for inhibiting diseases associated with estrogen deficiency or abnormal physiological response to endogenous estrogen
JP2004508373A (en) Cyclic and acyclic amidines and pharmaceutical compositions containing them for use as progesterone receptor binding agents
JPS60246315A (en) Selective d-2 dopamine receptor drug

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase