WO2015048311A1 - Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition - Google Patents

Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition Download PDF

Info

Publication number
WO2015048311A1
WO2015048311A1 PCT/US2014/057490 US2014057490W WO2015048311A1 WO 2015048311 A1 WO2015048311 A1 WO 2015048311A1 US 2014057490 W US2014057490 W US 2014057490W WO 2015048311 A1 WO2015048311 A1 WO 2015048311A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
oxy
imidazol
phenyl
dioxan
Prior art date
Application number
PCT/US2014/057490
Other languages
French (fr)
Inventor
Benjamin Eric Blass
Magid A Abou-Gharbia
Wayne E Childers
Pravin Iyer
Joshodeep BORUWA
Ramreddy BOBBALA
Rajashekar Reddy NIMMAREDDY
Original Assignee
Cortendo Ab (Publ)
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 Cortendo Ab (Publ) filed Critical Cortendo Ab (Publ)
Priority to CA2925294A priority Critical patent/CA2925294A1/en
Priority to US15/024,766 priority patent/US20160244436A1/en
Priority to EP14847772.2A priority patent/EP3049084A4/en
Priority to CN201480063953.0A priority patent/CN105764512A/en
Priority to JP2016516909A priority patent/JP2016536273A/en
Publication of WO2015048311A1 publication Critical patent/WO2015048311A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/46Drugs for disorders of the endocrine system of the suprarenal hormones for decreasing, blocking or antagonising the activity of glucocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • Q is selected from a group consisting of optionally substituted aryl, optionally substituted
  • R la , R lb , R lc , R ld , and R le are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C 3 -7 cycloalkyl, optionally substituted Ci_ 6 haloalkyl, C 1-6 , optionally substituted alkoxy, -NR 4a R 4b , -NR 5 COR 6 , -
  • R 2a , R 2b , R 2c , R 2d , R 2e , R 2f and R 2g are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C 3 -7 cycloalkyl, optionally substituted Ci_ 6 haloalkyl, Ci_ 6 optionally substituted alkoxy, -NR 4a R 4b , -NR 5 COR 6 , -
  • R 3 is selected from a group consisting of hydrogen, -S0 2 R 8 , -C(0)NR 9 R 10 ,
  • R a and R are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 5 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 6 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 7 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 8 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted C e haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl;
  • R 9 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7
  • R is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted C e branched alkyl;
  • R l la and R l lb are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH 2 OR 6 , and CE Heteroaryl.
  • compositions comprising an effective amount of at least one compounds according to the embodiments described in this document and at least one excipient.
  • Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve overproduction of Cortisol, including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the disease that involves overproduction of Cortisol is treated, delayed, slowed, or inhibited.
  • diseases that involve overproduction of Cortisol including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve overproduction of Cortisol, including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • diseases that involve overproduction of Cortisol, including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas
  • said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases or conditions associated with metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, or diseases that involve overproduction of Cortisol.
  • Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
  • Some embodiments relate to a method of modulating Cortisol activity, the method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the compound or composition modulates Cortisol. In some embodiments, the compound or composition lowers Cortisol levels in the subject.
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas and diseases that involve overproduction of Cortisol, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with overproduction of Cortisol. Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with overproduction of Cortisol, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments yet further relate to a method of lowering the concentration of Cortisol in the circulatory system.
  • Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
  • Some embodiments yet further relate to a method of lowering the concentration of Cortisol in the circulatory system, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl7 activity, including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the disease that involves excess Cypl7 activity is treated, delayed, slowed, or inhibited.
  • diseases that involve excess Cypl7 activity including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids
  • Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl7 activity, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to the embodiments described herein and an excipient.
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl7 activity, including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered, and wherein the disease that is associated with Cypl7 activity is treated, delayed, slowed, or inhibited.
  • diseases associated with Cypl7 activity including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer,
  • Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl7 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein Cypl7 activity is lowered.
  • Some embodiments also relate to a method for lowering Cypl7 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered.
  • lowering of Cypl7 activity leads to a lowering of testosterone levels to castrate levels in the subject.
  • lowering of Cypl7 activity leads to a lowering of estrogen levels to post- menopausal levels in the subject.
  • Some embodiments are directed to a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments described herein, wherein Cypl7 activity is lowered.
  • Cypl7 activity is inhibited almost completely or completely. In some embodiments, lowering of Cypl7 activity leads to a lowering of testosterone levels to castrate levels in the subject. In some embodiments, lowering of Cypl7 activity leads to a lowering of estrogen levels to post- menopausal levels in the subject.
  • Some embodiments relate to a method of lowering Cypl7 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl lBl activity, including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the disease that involves excess Cypl lB l activity is treated, delayed, slowed, or inhibited.
  • diseases that involve excess Cypl lBl activity including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsut
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl lBl activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (
  • Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl lBl activity, including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the Cypl lB activity is lowered and wherein the disease that involves excess Cypl 1B1 activity is treated, delayed, slowed, or inhibited.
  • diseases associated with Cypl lBl activity including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy,
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl lBl activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein the Cypl 1B1 activity is lowered.
  • diseases associated with Cypl lBl activity including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness,
  • Some embodiments also relate to a method for lowering Cypl lBl activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl 7 activity is lowered.
  • Some embodiments relate to a method of lowering Cy l 1B1 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments relate to a method of inhibiting Cypl lBl activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the disease that involves excess Cyp21 activity is treated, delayed, slowed, or inhibited.
  • diseases that involve excess Cyp21 activity including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycy
  • Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein Cyp21 activity is lowered, and wherein the disease that is associated with Cyp21 activity is treated, delayed, slowed, or inhibited.
  • diseases associated with Cyp21 activity including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), and
  • Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein the Cyp21 activity is lowered.
  • androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibr
  • Some embodiments also relate to a method for lowering Cyp21 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered. Some embodiments relate to a method of lowering Cyp21 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments relate to a method of inhibiting Cyp21 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
  • Some embodiments also relate to a method for lowering at least two of the following: Cypl7 activity, Cypl lBl activity, and Cyp21 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments described herein. In some embodiments, the method further modulates Cortisol.
  • Some embodiments relate to a method of treating, delaying, slowing, or inhibiting the progression of a disease selected from metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, related conditions, or a combination thereof, the method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the compound or composition lowers at least two of the following: Cypl7 activity, Cypl lBl activity, and Cyp21 activity in the subject.
  • the compound or composition modulates Cortisol.
  • the compound or composition lowers Cypl7 activity, Cypl lBl activity, and Cyp21 activity in the subject.
  • Some embodiments further relate to a process for preparing the compounds of embodiments described herein.
  • Embodiments of the present invention describe novel compounds useful for the treatment of diseases associated with the production of Cortisol, such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, and related conditions.
  • diseases that involve production of Cortisol comprise diseases that involve an overproduction of Cortisol.
  • diabetes mellitus includes diabetes mellitus type I, diabetes mellitus type II, prediabetes, latent autoimmune diabetes of adults (LAD A), congenital diabetes, cystic fibrosis-related diabetes, steroid diabetes, monogenic diabetes, gestational diabetes, or a combination thereof.
  • LAD A latent autoimmune diabetes of adults
  • congenital diabetes cystic fibrosis-related diabetes
  • steroid diabetes monogenic diabetes
  • gestational diabetes or a combination thereof.
  • Cortisol is a principal human glucocorticoid exhibiting many important physiological functions. It is involved in the regulation of the metabolism of proteins, carbohydrates, and fats; it counteracts insulin, maintains blood pressure and cardiovascular function, and suppresses the immune system's inflammatory response. However, pathological changes in adrenal and the upstream regulating switches can cause an overproduction of Cortisol.
  • One disease associated with overproduction of Cortisol is metabolic syndrome. Over the course of the last three decades, a growing body of knowledge has been developed to describe metabolic syndrome, also referred to as "Syndrome X" or "Insulin Resistance Syndrome” (Reaven, G. M. Role of insulin resistance in human disease, Diabetes, 1988, 37, 1595-1607).
  • Metabolic syndrome is defined as a cluster of abnormalities that occur in concert, including high blood pressure (BP), hyperglycemia, reduced high density lipoprotein cholesterol (HDL-C) levels, elevated triglycerides (TG) and abdominal obesity.
  • BP blood pressure
  • HDL-C reduced high density lipoprotein cholesterol
  • TG elevated triglycerides
  • NCEP National Cholesterol Education Program
  • ATP-III Adult Treatment Panel- ⁇
  • Cortisol production is regulated by several factors, including the enzymatic activity of the ⁇ ⁇ ⁇ -hydroxylase (Cypl lBl), 17a-hydroxylase-C17,20-lyase (Cypl7), and 21 -hydroxylase (Cyp21). All three are members of the cytochrome P450 superfamily of enzymes.
  • the 17a-hydroxylase/Ci7_2o lyase enzyme complex is essential for the biosynthesis of androgens.
  • CYP17 is a bifunctional enzyme which possess both a C 17-2 o- lyase activity and a C17-hydroxylase activity.
  • Cypl lB l catalyzes the final step of Cortisol synthesis, hydroxylation of the C-l l position of deoxycortisol.
  • Cypl7 has multiple functions in corticosteroid synthesis. The C-17 and C-20 positions of the steroid framework can be modified by this enzyme. Pregnenolone and progesterone are hydroxylated by Cypl7 at C-17 (hydroxylase activity), while the C-20/C-17 bond is cleaved by the same enzyme in 17-hydroxyprogesterone and 17-hydroxypregnenolone (lyase activity).
  • Cyp21 catalyzes the hydroxylation of C-21 in steroids such as progesterone and 17a-hydroxy progesterone.
  • Cortisol compounds that are dual inhibitors of Cypl7 and Cyp21 will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • Cortisol compounds that are dual inhibitors of Cypl7 and Cypl lBl will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • Cortisol compounds that are dual inhibitors of Cypl lBl and Cyp21 will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • Cortisol diseases and symptoms associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas , that are both disease- modifying and effective in treating patients.
  • diseases and symptoms associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas , that are both disease- modifying and effective in treating patients.
  • Embodiments of the present invention addresses the need to identify effective treatment for diseases and symptoms associated with the overproduction of Cortisol, such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • diseases and symptoms associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • Cortisol lowering agents of embodiments described herein are capable of treating, delaying, slowing, or inhibiting the progression of diseases associated with the overproduction of Cortisol such as, for example, metabolic syndrome.
  • Cortisol is a principal human glucocorticoid exhibiting many important physiological functions. It is involved in the regulation of the metabolism of proteins, carbohydrates, and fats; it counteracts insulin, maintains blood pressure and cardiovascular function, and suppresses the immune system's inflammatory response.
  • pathological changes in adrenal gland or other tissues capable of secreting Cortisol and the upstream regulating switches can cause an overproduction of Cortisol.
  • One disease associated with overproduction of Cortisol is metabolic syndrome.
  • Cortisol is associated with hypertension, diabetes mellitus, obesity, headache, depression, Cushing's syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • Cortisol lowering agents of embodiments described in this disclosure ameliorate, abate, otherwise cause to be controlled, diseases associated with the overproduction of Cortisol, for example metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • diseases associated with the overproduction of Cortisol for example metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
  • compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present teachings also consist essentially of, or consist of, the recited components, and that the processes of the present teachings also consist essentially of, or consist of, the recited processing steps.
  • the term “consisting essentially of or “consists essentially of” means that the only active pharmaceutical ingredient in the formulation or method that treats the specified condition (e.g. Cushing's syndrome) is the specifically recited active pharmaceutical ingredient for treating the specified condition in the particular embodiment or claim; that is, the scope of the claim or embodiment is limited to the specified elements or steps and those that do not materially affect the basic and novel characteristic(s) of the particular embodiment or claimed invention.
  • excess Cypl7 activity may refer to an above normal level of the C17-hydroxylase activity of CYP17 which promotes the overproduction of glucocorticoids or an above normal level of the C17,20-lyase activity of Cypl7 which promotes the overproduction of sex hormones. In some embodiments, excess Cypl7 activity may lead to overproduction of Cortisol or an overproduction of androgenic or estrogenic hormones [0059] As used herein, the term "halogen” includes chlorine, bromine, fluorine, iodine, or a combination thereof.
  • alkyl and/or “aliphatic” whether used alone or as part of a substituent group refers to straight and branched carbon chains having 1 to 20 carbon atoms or any number within this range, for example 1 to 6 carbon atoms or 1 to 4 carbon atoms.
  • Designated numbers of carbon atoms e.g. C ⁇ refers independently to the number of carbon atoms in an alkyl moiety or to the alkyl portion of a larger alkyl-containing substituent.
  • alkyl groups include methyl, ethyl, n-propyl, z ' so-propyl, n-butyl, sec-butyl, z ' so-butyl, tert-butyl, and the like.
  • Alkyl groups can be optionally substituted.
  • substituted alkyl groups include hydroxymethyl, chloromethyl, trifluoromethyl, aminomethyl, 1- chloroethyl, 2-hydroxyethyl, 1,2-difluoroethyl, 3-carboxypropyl, and the like.
  • substituent groups with multiple alkyl groups such as (Ci_ 6 alkyl) 2 amino, the alkyl groups may be the same or different.
  • alkenyl and alkynyl groups refer to straight and branched carbon chains having 2 or more carbon atoms, preferably 2 to 20, wherein an alkenyl chain has at least one double bond in the chain and an alkynyl chain has at least one triple bond in the chain.
  • Alkenyl and alkynyl groups can be optionally substituted.
  • Non-limiting examples of alkenyl groups include ethenyl, 3-propenyl, 1-propenyl (also 2-methylethenyl), isopropenyl (also 2-methylethen-2-yl), buten-4-yl, and the like.
  • Non-limiting examples of substituted alkenyl groups include 2-chloroethenyl (also 2-chlorovinyl), 4-hydroxybuten-l-yl, 7- hydroxy-7-methyloct-4-en-2-yl, 7-hydroxy-7-methyloct-3,5-dien-2-yl, and the like.
  • Non-limiting examples of alkynyl groups include ethynyl, prop-2-ynyl (also propargyl), propyn-l-yl, and 2-methyl-hex-4-yn-l-yl.
  • Non-limiting examples of substituted alkynyl groups include, 5-hydroxy-5-methylhex-3-ynyl, 6-hydroxy-6-methylhept-3-yn-2-yl, 5- hydroxy-5-ethylhept-3-ynyl, and the like.
  • cycloalkyl refers to a non-aromatic carbon-containing ring including cyclized alkyl, alkenyl, and alkynyl groups, e.g., having from 3 to 14 ring carbon atoms, preferably from 3 to 7 or 3 to 6 ring carbon atoms, or even 3 to 4 ring carbon atoms, and optionally containing one or more (e.g., 1, 2, or 3) double or triple bond.
  • cycloalkyl groups may be monocyclic (e.g., cyclohexyl) or polycyclic (e.g., containing fused, bridged, and/or spiro ring systems), wherein the carbon atoms are located inside or outside of the ring system. Any suitable ring position of the cycloalkyl group can be covalently linked to the defined chemical structure. In some embodiments, cycloalkyl rings may be optionally substituted.
  • Non-limiting examples of cycloalkyl groups include: cyclopropyl, 2-methyl-cyclopropyl, cyclopropenyl, cyclobutyl, 2,3-dihydroxycyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctanyl, decalinyl, 2,5-dimethylcyclopentyl, 3,5-dichlorocyclohexyl, 4- hydroxycyclohexyl, 3,3,5-trimethylcyclohex-l-yl, octahydropentalenyl, octahydro-lH- indenyl, 3a,4,5,6,7,7a-hexahydro-3H-inden-4-yl, decahydroazulenyl; bicyclo[6.2.0]de
  • cycloalkyl also includes carbocyclic rings which are bicyclic hydrocarbon rings, non- limiting examples of which include, bicyclo-[2.1.1]hexanyl, bicyclo[2.2.1]heptanyl, bicyclo[3.1.1]heptanyl, l,3-dimethyl[2.2.1]heptan-2-yl, bicyclo[2.2.2]octanyl, and bicyclo [3.3.3 ]undecanyl.
  • haloalkyl may include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen.
  • Haloalkyl groups include perhaloalkyl groups, wherein all hydrogens of an alkyl group have been replaced with halogens (e.g., -CF 3 , -CF 2 CF 3 ).
  • Haloalkyl groups can optionally be substituted with one or more substituents in addition to halogen.
  • haloalkyl groups include, but are not limited to, fluoromethyl, dichloroethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl groups.
  • alkoxy refers to the group -O-alkyl, wherein the alkyl group is as defined above. Alkoxy groups optionally may be substituted.
  • C3-C6 cyclic alkoxy refers to a ring containing 3 to 6 carbon atoms and at least one oxygen atom (e.g., tetrahydrofuran, tetrahydro-2H-pyran). C3-C6 cyclic alkoxy groups optionally may be substituted.
  • aryl wherein used alone or as part of another group, is defined herein as a an unsaturated, aromatic monocyclic ring of 6 carbon members or to an unsaturated, aromatic polycyclic ring of from 10 to 14 carbon members.
  • Aryl rings can be, for example, phenyl or naphthyl ring each optionally substituted with one or more moieties capable of replacing one or more hydrogen atoms.
  • Non-limiting examples of aryl groups include: phenyl, naphthylen-l-yl, naphthylen-2-yl, 4-fluorophenyl, 2-hydroxyphenyl, 3- methylphenyl, 2-amino-4-fluorophenyl, 2-(N,N-diethylamino)phenyl, 2-cyanophenyl, 2,6- di-tert-butylphenyl, 3-methoxyphenyl, 8-hydroxynaphthylen-2-yl 4,5- dimethoxynaphthylen-l-yl, and 6-cyano-naphthylen-l-yl.
  • Aryl groups also include, for example, phenyl or naphthyl rings fused with one or more saturated or partially saturated carbon rings (e.g., bicyclo[4.2.0]octa-l,3,5-trienyl, indanyl), which can be substituted at one or more carbon atoms of the aromatic and/or saturated or partially saturated rings.
  • phenyl or naphthyl rings fused with one or more saturated or partially saturated carbon rings (e.g., bicyclo[4.2.0]octa-l,3,5-trienyl, indanyl), which can be substituted at one or more carbon atoms of the aromatic and/or saturated or partially saturated rings.
  • arylalkyl refers to the group -alkyl-aryl, where the alkyl and aryl groups are as defined herein.
  • Aralkyl groups of embodiments described herein are optionally substituted. Examples of arylalkyl groups include, for example, benzyl, 1 -phenylethyl, 2-phenylethyl, 3-phenylpropyl, 2-phenylpropyl, fluorenylmethyl and the like.
  • heterocyclic and/or “heterocycle” and/or “heterocylyl,” whether used alone or as part of another group, are defined herein as one or more ring having from 3 to 20 atoms wherein at least one atom in at least one ring is a heteroatom selected from nitrogen ( ⁇ ), oxygen (O), or sulfur (S), and wherein further the ring that includes the heteroatom is non-aromatic.
  • the non-heteroatom bearing ring may be aryl (e.g., indolinyl, tetrahydroquinolinyl, chromanyl).
  • heterocycle groups have from 3 to 14 ring atoms of which from 1 to 5 are heteroatoms independently selected from nitrogen ( ⁇ ), oxygen (O), or sulfur (S).
  • nitrogen
  • O oxygen
  • S sulfur
  • One or more ⁇ or S atoms in a heterocycle group can be oxidized.
  • Heterocycle groups can be optionally substituted.
  • Non-limiting examples of heterocyclic units having a single ring include: diazirinyl, aziridinyl, urazolyl, azetidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolidinyl, isothiazolyl, isothiazolinyl oxathiazolidinonyl, oxazolidinonyl, hydantoinyl, tetrahydrofuranyl, pyrrolidinyl, morpholinyl, piperazinyl, piperidinyl, dihydropyranyl, tetrahydropyranyl, piperidin-2-onyl (valerolactam), 2,3,4,5- tetrahydro-lH-azepinyl, 2,3-dihydro-lH-indole, and 1,2,3,4-
  • Non- limiting examples of heterocyclic units having 2 or more rings include: hexahydro-lH- pyrrolizinyl, 3a,4,5,6,7,7a-hexahydro-lH-benzo[d]imidazolyl, 3a,4,5,6,7,7a-hexahydro- lH-indolyl, 1,2,3,4-tetrahydroquinolinyl, chromanyl, isochromanyl, indolinyl, isoindolinyl, and decahydro-lH-cycloocta[b]pyrrolyl.
  • heteroaryl whether used alone or as part of another group, is defined herein as one or more rings having from 5 to 20 atoms wherein at least one atom in at least one ring is a heteroatom chosen from nitrogen (N), oxygen (O), or sulfur (S), and wherein further at least one of the rings that includes a heteroatom is aromatic.
  • the non-heteroatom bearing ring may be a carbocycle (e.g., 6,7-Dihydro-5H-cyclopentapyrimidine) or aryl (e.g., benzofuranyl, benzothiophenyl, indolyl).
  • heteroaryl groups have from 5 to 14 ring atoms and contain from 1 to 5 ring heteroatoms independently selected from nitrogen (N), oxygen (O), or sulfur (S). One or more N or S atoms in a heteroaryl group can be oxidized. Heteroaryl groups can be substituted.
  • heteroaryl rings containing a single ring include: 1,2,3,4-tetrazolyl, [l,2,3]triazolyl, [l,2,4]triazolyl, triazinyl, thiazolyl, lH-imidazolyl, oxazolyl, furanyl, thiopheneyl, pyrimidinyl, 2- phenylpyrimidinyl, pyridinyl, 3-methylpyridinyl, and 4-dimethylaminopyridinyl.
  • Non- limiting examples of heteroaryl rings containing 2 or more fused rings include: benzofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, cinnolinyl, naphthyridinyl, phenanthridinyl, 7H-purinyl, 9H-purinyl, 6-amino-9H-purinyl, 5H- pyrrolo[3,2-( Jpyrimidinyl, 7H-pyrrolo[2,3-( Jpyrimidinyl, pyrido[2,3- ⁇ i]pyrimidinyl, 2- phenylbenzo[d]thiazolyl, lH-indolyl, 4,5,6,7-tetrahydro-l-H-indolyl, quinoxalinyl, 5- methylquinoxalinyl, quinazolinyl, quinolinyl, 8-hydroxy-quinolinyl, and iso
  • heteroaryl group as described above is C 1 -C5 heteroaryl, which has 1 to 5 carbon ring atoms and at least one additional ring atom that is a heteroatom (preferably 1 to 4 additional ring atoms that are heteroatoms) independently selected from nitrogen (N), oxygen (O), or sulfur (S).
  • N nitrogen
  • O oxygen
  • S sulfur
  • C 1 -C5 heteroaryl examples include, but are not limited to, triazinyl, thiazol-2-yl, thiazol-4-yl, imidazol-l-yl, 1H- imidazol-2-yl, lH-imidazol-4-yl, isoxazolin-5-yl, furan-2-yl, furan-3-yl, thiophen-2-yl, thiophen-4-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridin-2-yl, pyridin-3-yl, and pyridin-4-yl.
  • the ring when two substituents are taken together to form a ring having a specified number of ring atoms (e.g., R 2 and R 3 taken together with the nitrogen (N) to which they are attached to form a ring having from 3 to 7 ring members), the ring can have carbon atoms and optionally one or more (e.g., 1 to 3) additional heteroatoms independently selected from nitrogen (N), oxygen (O), or sulfur (S).
  • the ring can be saturated or partially saturated and can be optionally substituted.
  • a fused ring unit contains heteroatoms in both a saturated and an aryl ring
  • the aryl ring will predominate and determine the type of category to which the ring is assigned. For example, l,2,3,4-tetrahydro-[l ,8]naphthyridine having the formula:
  • substituted is used throughout the specification.
  • substituted is defined herein as a moiety, whether acyclic or cyclic, which has one or more hydrogen atoms replaced by a substituent or several (e.g., 1 to 10) substituents as defined herein below.
  • the substituents are capable of replacing one or two hydrogen atoms of a single moiety at a time.
  • these substituents can replace two hydrogen atoms on two adjacent carbons to form said substituent, new moiety or unit.
  • a substituted unit that requires a single hydrogen atom replacement includes halogen, hydroxyl, and the like.
  • a two hydrogen atom replacement includes carbonyl, oximino, and the like.
  • a two hydrogen atom replacement from adjacent carbon atoms includes epoxy, and the like.
  • substituted is used throughout the present specification to indicate that a moiety can have one or more of the hydrogen atoms replaced by a substituent. When a moiety is described as “substituted” any number of the hydrogen atoms may be replaced.
  • difluoromethyl is a substituted Ci alkyl
  • trifluoromethyl is a substituted Ci alkyl
  • 4-hydroxyphenyl is a substituted aromatic ring
  • (N,N-dimethyl-5-amino)octanyl is a substituted Cs alkyl
  • 3-guanidinopropyl is a substituted C 3 alkyl
  • 2-carboxypyridinyl is a substituted heteroaryl.
  • variable groups defined herein e.g., alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryloxy, aryl, heterocycle and heteroaryl groups defined herein, whether used alone or as part of another group, can be optionally substituted. Optionally substituted groups will be so indicated.
  • the substituents are selected from:
  • -OR 14 for example, -OH, -OCH3, -OCH 2 CH 3 , -OCH 2 CH 2 CH 3 ;
  • -C(0)OR 14 for example, -C0 2 CH 3 , -C0 2 CH 2 CH 3 , -C0 2 CH 2 CH 2 CH 3 ; iv) -C(0)N(R 14 ) 2 ; for example, -CONH 2 , -CONHCH3, -CON(CH 3 ) 2 ;
  • -SO2R 14 for example, -S0 2 H; -S0 2 CH :
  • each R is independently hydrogen, optionally substituted C1-C6 linear or branched alkyl (e.g., optionally substituted C1-C4 linear or branched alkyl), or optionally substituted C3-C6 cycloalkyl (e.g optionally substituted C3-C4 cycloalkyl); or two R 14 units can be taken together to form a ring comprising 3-7 ring atoms.
  • each R 14 is independently hydrogen, C1-C6 linear or branched alkyl optionally substituted with halogen or C3-C6 cycloalkyl or C3-C6 cycloalkyl.
  • Ci_6 alkyl is specifically intended to individually disclose Ci, C 2 , C3, C 4 , C5, Ce, Ci-Ce, C1-C5, C -C4, C -C3, C1-C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C 4 -C 6 , C4-C5, and C 5 -C 6 , alkyl.
  • Compounds described herein can contain an asymmetric atom (also referred as a chiral center), and some of the compounds can contain one or more asymmetric atoms or centers, which can thus give rise to optical isomers (enantiomers) and diastereomers.
  • asymmetric atom also referred as a chiral center
  • the present teachings and compounds disclosed herein include such enantiomers and diastereomers, as well as the racemic and resolved, enantiomerically pure R and S stereoisomers, as well as other mixtures of the R and S stereoisomers and pharmaceutically acceptable salts thereof.
  • Optical isomers can be obtained in pure form by standard procedures known to those skilled in the art, which include, but are not limited to, diastereomeric salt formation, kinetic resolution, and asymmetric synthesis.
  • the present teachings also encompass cis and trans isomers of compounds containing alkenyl moieties (e.g., alkenes and imines). It is also understood that the present teachings encompass all possible regioisomers, and mixtures thereof, which can be obtained in pure form by standard separation procedures known to those skilled in the art, and include, but are not limited to, column chromatography, thin-layer chromatography, and high-performance liquid chromatography.
  • compositions of the present teachings which can have an acidic moiety, can be formed using organic and inorganic bases. Both mono and polyanionic salts are contemplated, depending on the number of acidic hydrogens available for deprotonation.
  • Suitable salts formed with bases include metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, or magnesium salts; ammonia salts and organic amine salts, such as those formed with morpholine, thiomorpholine, piperidine, pyrrolidine, a mono-, di- or tri-lower alkylamine (e.g., ethyl- tert-butyl-, diethyl-, diisopropyl-, triethyl-, tributyl- or dimethylpropylamine), or a mono-, di-, or trihydroxy lower alkylamine (e.g., mono-, di- or triethanolamine).
  • metal salts such as alkali metal or alkaline earth metal salts, for example sodium, potassium, or magnesium salts
  • ammonia salts and organic amine salts such as those formed with morpholine, thiomorpholine, piperidine, pyrrolidine, a mono-
  • inorganic bases include aHC0 3 , a 2 C0 3 , KHCO 3 , K2CO 3 , CS2CO3, LiOH, NaOH, KOH, NaH 2 P0 4 , Na 2 HP0 4 , and Na 3 P0 4 .
  • Internal salts also can be formed.
  • salts can be formed using organic and inorganic acids.
  • salts can be formed from the following acids: acetic, propionic, lactic, benzenesulfonic, benzoic, camphorsulfonic, citric, tartaric, succinic, dichloroacetic, ethenesulfonic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, malonic, mandelic, methanesulfonic, mucic, napthalenesulfonic, nitric, oxalic, pamoic, pantothenic, phosphoric, phthalic, propionic, succinic, sulfuric, tartaric, toluenesulfonic, and camphorsulfonic as well as other known pharmaceutically acceptable acids.
  • any variable occurs more than one time in any constituent or in any formula, its definition in each occurrence is independent of its definition at every other occurrence (e.g., in N(R 13 )2, each R 13 may be the same or different than the other). Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • the terms "treat” and “treating” and “treatment” as used herein, refer to partially or completely alleviating, inhibiting, ameliorating and/or relieving a condition from which a patient is suspected to suffer.
  • terapéuticaally effective and “effective dose” refer to a substance or an amount that elicits a desirable biological activity or effect.
  • a "therapeutically effective amount” or “effective amount” of a composition is a predetermined amount calculated to achieve the desired effect, i.e. treat, delay, slow, or inhibit the progression of diseases that involve overproduction of Cortisol.
  • the activity contemplated by the present methods includes both medical therapeutic and/or prophylactic treatment, as appropriate.
  • the specific dose of a compound administered according to this invention to obtain therapeutic and/or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the compound administered, the route of administration, and the condition being treated.
  • the compounds are effective over a wide dosage range and, for example, dosages per day will normally fall within the range of from 0.001 to 10 mg/kg, more usually in the range of from 0.01 to 1 mg/kg.
  • a therapeutically effective amount of compound of this invention is typically an amount such that when it is administered in a physiologically tolerable excipient composition, it is sufficient to achieve an effective systemic concentration or local concentration in the tissue.
  • the terms “subject” or “patient” are used interchangeably and refer to mammals such as human patients and non-human primates, as well as experimental animals such as rabbits, rats, and mice, and other animals. Accordingly, the term “subject” or “patient” as used herein means any mammalian patient or subject to which the compounds of the invention can be administered.
  • accepted screening methods are employed to determine risk factors associated with a targeted or suspected disease or condition or to determine the status of an existing disease or condition in a subject. These screening methods include, for example, conventional work-ups to determine risk factors that may be associated with the targeted or suspected disease or condition. These and other routine methods allow the clinician to select patients in need of therapy using the methods and compounds of embodiments described herein.
  • Q is selected from a group consisting of optionally substituted aryl, optionally substituted heteroaryl,
  • R la , R lb , R lc , R ld , and R le are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_ 6 haloalkyl, C 1-6 , optionally substituted alkoxy, -NR 4a R 4b , -NR 5 COR 6 , -
  • R 2a , R 2b , R 2c , R 2d , R 2e , R 2f and R 2g are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_ 6 haloalkyl, Ci_ 6 optionally substituted alkoxy, -NR 4a R 4b , -NR 5 COR 6 , -
  • R 3 is selected from a group consisting of hydrogen, -S0 2 R 8 , -C(0)NR 9 R 10 , -
  • R 4a and R 4b are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 5 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 6 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 7 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
  • R 8 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted C e haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl;
  • R 9 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7
  • R is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted C e branched alkyl;
  • R l la and R l lb are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH 2 OR 6 , and CH 2 Heteroaryl.
  • Some embodiments include com ounds having formula (II):
  • Some embodiments include compounds having formula (III):
  • Some embodiments include com ounds having formula (IV):
  • Some embodiments include compounds having formula (V):
  • Some embodiments include compounds having formula (VI):
  • Some embodiments include compounds having formula (VII):
  • Some embodiments include compounds having formula (VIII):
  • inventions include compounds having formula (IX):
  • Some embodiments include compounds having formula (X):
  • Some embodiments include compounds having formula (Xa):
  • Q is optionally substituted aryl.
  • Q is optionally substituted heteroaryl.
  • Q is ⁇ — / .
  • R la of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , S0 2 R 7 and - S0 2 NHR 6 .
  • R lb of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R lc of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R ld of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci-6 haloalkyl, Ci-6, optionally substituted alkoxy, - NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R le of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2a of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2b of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2c of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2d of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2e of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2f of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 2g of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy,
  • R 2a is -NR 4a R 4b , -NR 5 COR 6 , -C0 2 R 6 , -C0 2 NR 4a R 4b , -NHS0 2 R 7 , -SH, -SR 7 , -S0 2 R 7 , and - S0 2 NHR 6 .
  • R 3 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, -S0 2 R 8 , -C(0)NR 9 R 10 , -C(0)R 7 , -
  • R 4a of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
  • R 4b of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
  • R 5 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alky, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
  • R 6 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci-6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
  • R 7 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl.
  • R 8 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl.
  • R 9 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci-6 haloalkyl,
  • R 10 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted Ci-6 branched alkyl.
  • R lla of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH 2 OR 6 , and CH 2 Heteroaryl.
  • R l lb of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH 2 OR 6 , and CH 2 Heteroaryl.
  • Exemplary embodiments include compounds having the formula (X) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , and R 3 are defined herein below in Table 2.
  • Exemplary embodiments include compounds having the formula (XII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , and R 3 are defined herein below in Table 3.
  • Exemplary embodiments include compounds having the formula (XIII) or a pharmaceutically acceptable salt form thereof:
  • Exemplary embodiments include compounds having the formula (XIV) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , R 9 , and R 10 are defined herein below in Table 5.
  • Exemplary embodiments include compounds having the formula (XV) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , R l la , R l lb , and R 7 are defined herein below in Table 6.
  • Exemplary embodiments include compounds having the formula (XVI) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , R l la , R l lb , and R 7 are defined herein below in Table 7.
  • Exemplary embodiments include compounds having the formula (XVII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , R lla , R llb , R 4a , and R 4b are defined herein below in Table 8.
  • R la , R lb , R lc , R ld , R le , R l la , R l lb , R 4a , and R 4b are defined herein below in Table 9.
  • Exemplary embodiments include compounds having the formula (XIX) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , and R 7 are defined herein below in Table 10.
  • Exemplary embodiments include compounds having the formula (XX) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le , and R 7 are defined herein below in Table 1 1.
  • R la , R lb , R lc , R ld , and R le are defined herein below in
  • Exemplary embodiments include compounds having the formula (XXII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , and R le are defined herein below in
  • Exemplary embodiments include compounds having the formula (XXIII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , and R le are defined herein below in Table 14.
  • Exemplary embodiments include compounds having the formula (XXIV) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , and R le are defined herein below in Table 15.
  • Exemplary embodiments include compounds having the formula (XXV) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , and R le are defined herein below in Table 16.
  • Exemplary embodiments include compounds having the formula (XXVI) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , and R le are defined herein below in Table 17.
  • Exemplary embodiments include compounds having the formula (XXVII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le and R 7 are defined herein below in Table 18.
  • Exemplary embodiments include compounds having the formula (XXVIII) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le and R 7 are defined herein below in Table 19.
  • Exemplary embodiments include compounds having the formula (XXVIIIa) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le and R 7 are defined herein below in Table 20.
  • Exemplary embodiments include compounds having the formula (XXVIIIb) or a pharmaceutically acceptable salt form thereof:
  • R la , R lb , R lc , R ld , R le and R 7 are defined herein below in Table 21.
  • the compound having the formula (XXXIV) (XXXIV) has the chemical name methyl 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazine- 1 -carboxylate.
  • Some embodiments of the present invention further relate to a process for preparing the Cortisol lowering agents of embodiments described herein.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., l H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high pressure liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
  • HPLC high pressure liquid chromatograpy
  • GC gas chromatography
  • GPC gel-permeation chromatography
  • TLC thin layer chromatography
  • Preparation of the compounds can involve protection and deprotection of various chemical groups.
  • the need for protection and deprotection and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Greene et al, Protective Groups in Organic Synthesis, 2d. Ed. (Wiley & Sons, 1991), the entire disclosure of which is incorporated by reference herein for all purposes.
  • Suitable solvents typically are substantially nonreactive with the reactants, intermediates, and/or products at the temperatures at which the reactions are carried out, i.e., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • the compounds of these teachings can be prepared by methods known in the art of organic chemistry.
  • the reagents used in the preparation of the compounds of these teachings can be either commercially obtained or can be prepared by standard procedures described in the literature.
  • compounds of embodiments described herein can be prepared according to the method illustrated in the General Synthetic Schemes.
  • the reagents used in the preparation of the compounds of this invention can be either commercially obtained or can be prepared by standard procedures described in the literature.
  • compounds in the genus may be produced by one of the following reaction schemes.
  • a suitably substituted compound of formula (1) a known compound or compound prepared by known methods, is reacted with a bromine in an organic solvent such as 1 ,4- dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, N,N- dimethylformamide, and the like to provide a compound of the formula (2).
  • an organic solvent such as 1 ,4- dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, N,N- dimethylformamide, and the like.
  • a compound of the formula (2) is then reacted with trimethyl orthoacetate in the presence of an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like in a solvent such as methanol, 1,4-dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, and the like optionally with heating to provide a compound of the formula (3).
  • an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like
  • a solvent such as methanol, 1,4-dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, and the like optionally with heating to provide a compound of the formula (3).
  • a compound of the formula (3) is then reacted with a compound of the formula (4), a known compound or compound prepared by known methods, in the presence of an acid such as p-toluenesulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like in a solvent such as benzene, toluene, p- xylene, 1 ,4-dioxane, tetrahydrofuran, and the like to provide a compound of the formula (5).
  • an acid such as p-toluenesulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like
  • a solvent such as benzene, toluene, p- xylene, 1 ,4-dioxane, tetrahydrofuran, and the like to provide a compound of the formula (5).
  • a compound of the formula (5) is then reacted with a compound of the formula (6), a known compound or compound prepared by known methods, in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like, in a solvent such as N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide, 1,4-dioxane, tetrahydrofuran, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (7).
  • a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like
  • a solvent such as N,N-dimethylformamide, ⁇ , ⁇ -dimethylacetamide, 1,4-dioxane, tetrahydrofuran, optionally with heating, optionally with microwave irradi
  • Benzaldehyde is reacted with a glycerol in an organic solvent such as 1,4-dioxane, tetrahydrofuran, 1,2-dichloroethane, ⁇ , ⁇ -dimethylformamide, N,N-dimethylacetamide, and the like, in the presence of an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (9).
  • an organic solvent such as 1,4-dioxane, tetrahydrofuran, 1,2-dichloroethane, ⁇ , ⁇ -dimethylformamide, N,N-dimethylacetamide, and the like
  • an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid,
  • a compound of the formula (9) is then reacted with a compound of the formula (10) wherein X is a leaving group such as bromine, chlorine, methansulfonate, and the like, in the presence of a base such as sodium hydride, potassium hydride, lithium diisopropylamide, sodium diisopropylamide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and the like in an solvent such as 1,4-dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (11).
  • a base such as sodium hydride, potassium hydride, lithium diisopropylamide, sodium diisopropylamide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and the like
  • an solvent such as 1,4-di
  • a compound of the formula (11) is then reacted with an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (4).
  • an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (4).
  • a compound of the formula (7) is reacted with a compound of the formula (12), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium, and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xylene
  • a compound of the formula (13) is reacted with a base such as a potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as methanol, ethanol, isopropanol, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (14).
  • a base such as a potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like
  • a solvent such as methanol, ethanol, isopropanol, and the like
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (15), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (16).
  • a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like
  • organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ ,
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (17), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (18).
  • a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like
  • organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ ,
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (19), a known compound or compound prepared by known methods in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like to provide a compound of the formula (20).
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods is reacted with a p- nitrophenylchloroformate in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (21).
  • a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, and the like
  • organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylform
  • a compound of formula (21) is then reacted with a compound of the formula (22), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (20).
  • Scheme 9
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (23), in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine,
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (25), a known compound or compound prepared by known methods wherein n is 1 or 2, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1 ,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (26).
  • a compound of formula (26) is then reacted with a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, potassium carbonate, sodium carbonate, lithium carbonate, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like to provide a compound of the formula (27).
  • a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, potassium carbonate, sodium carbonate, lithium carbonate, and the like
  • organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like
  • a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (28), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, ⁇ , ⁇ -dimethylformamide, and the like to provide a compound of the formula (29).
  • a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like
  • organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane,
  • a compound of the formula (7) is reacted with a compound of the formula (30), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium, [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xylene
  • a compound of the formula (7) is reacted with a compound of the formula (32), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium, [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xy
  • Examples 1-X provide methods for preparing representative compounds of the disclosure The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare additional compounds of embodiments described herein.
  • Example 1 Synthesis of 2-Bromo-l-(2, 4-dichlorophenyl)ethanone.
  • Example 6 Synthesis of (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2- (2,4-dichlorophenyl)-l,3-dioxane and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)- 2-(2,4-dichlorophenyl)-l,3-dioxane.
  • the following compounds can be prepared by the procedure of (2s,5s)-5-((4- bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane and (2r,5r)-5- ((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane.
  • the skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
  • Example 7 Synthesis of (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)- 1 ,3 -dioxane.
  • Example 8 Synthesis of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- 1 H-imidazole.
  • Example 9 Synthesis of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2,4- dichlorophenyl)-l,3-dioxa -2-yl)methyl)-lH-imidazole (7b):
  • Example 10 Synthesis of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- lH-imidazole.
  • Example 1 1 Synthesis of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- lH-imidazole.
  • the crude product was purified by column chromatography on silica (100-200 mesh) eluting with 15% ethyl acetate in petroleum ether to give an oil.
  • the oil was dissolved in tetrahydrofuran (200 mL) and added dropwise over 45 minutes to a suspension of NaH (53.33 g, 2.22 mol) in tetrahydrofuran (800 mL).
  • 4- bromobenzylbromide (277.7 g, 1.1 1 mol) was added portionwise over 30 minutes and the reaction mixture was stirred at room temperature for 6 hours.
  • the reaction mixture was poured into ice-water and extracted with ethyl acetate.
  • Example 14 Synthesis of l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone
  • Example 15 Synthesis of l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone.
  • Example 16 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazine.
  • Example 17 Synthesis of 3-((4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l, -dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)sulfonyl)benzonitrile.
  • the reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na 2 S0 4 ) filtered and concentrated.
  • the crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound.
  • Example 18 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-((3 - chloropropyl)sulfonyl)piperazine.
  • the reaction mixture was poured into ice- water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na 2 S0 4 ) filtered and concentrated.
  • the crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound.
  • Example 19 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- lsulfonyl)piperazine.
  • the reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na 2 S0 4 ) filtered and concentrated.
  • the crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound.
  • Example 20 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- lfonyl)piperazine.
  • the reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried ( a 2 S04) filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound.
  • Example 22 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-(( 1 H-imidazol-4- yl)sulfonyl)piperazine.
  • the reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na 2 S0 4 ) filtered and concentrated.
  • the crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound.
  • Example 23 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-((3 - (trifluoromethoxy)phenyl)sulfonyl)piperazine.
  • Example 24 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- sulfonyl)piperazine.
  • the reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried ( a 2 S0 4 ) filtered and concentrated.
  • the crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound.
  • Example 26 Synthesis of l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)ethanone.
  • reaction mixture was refluxed over 2 hours.
  • the reaction mixture was filtered and concentrated and the residue was purified by column chromatography on silica (100-200 mesh) eluting with 10% methanol in ethyl acetate to afford the title compound.
  • Example 27 Synthesis of l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)ethanone.
  • Example 28 Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dio
  • Example 29 Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)morpholine.
  • Example 30 Synthesis of 4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)morpholine.
  • Example 31 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine:
  • Example 32 Synthesis of l-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine:
  • Example 33 ynthesis of 2-methoxy ethyl 4-(4-((((2s,5s)-2-((lH-imidazol-l- yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l- carboxylate:
  • Example 34 Synthesis of 2-methoxyethyl 4-(4-((((2r,5r)-2-((lH-imidazol-l- yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l- carboxylate:
  • Example 35 Synthesis of ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2- (2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate:
  • Example 36 Synthesis of ethyl 2-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2- (2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate:
  • Example 37 Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine- 1 - carboxamide.
  • Example 38 Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)pyridine.
  • Example 39 The compounds in the Table 23 below represent the extraction of over 200 compounds which realized the in vitro and in vivo goals.
  • the target goals are defined in Table 22.
  • the in vitro goals are defined by efficacy targets: CYP17, CYP11, and CYP21.
  • the off-target enzymes (where potency should be low) are CYP 19 and CYP3A4.
  • Other parameters are no liver effects also estimated by bile acid synthesis inhibition.
  • Table 23 Representative examples of compounds of the disclosure and their potencies in Cyp l7, Cypl I, and Cyp21 assays.
  • Example 40 Pharmacokinetic studies in the guinea pig were run using lmg/kg IV dosing (20%DMA, 40% TEG, 40% water) and 10 mg/kg oral dosing (2% Tween-80, 98% HPMC (1 % water)).
  • the oral PK data is summarized in the tables below.
  • compositions or formulations which comprise the Cortisol lowering agents according to embodiments described herein.
  • the compositions of embodiments described herein comprise an effective amount of one or more compounds of the disclosure and salts thereof according to embodiments described herein which are effective for providing Cortisol lowering; and one or more excipients.
  • excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means to achieve effective absorption by the recipient of the active ingredient.
  • An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach.
  • the formulator can also take advantage of the fact the compounds of embodiments described herein have improved cellular potency, pharmacokinetic properties, as well as improved oral bioavailability.
  • compositions that include at least one compound described herein and one or more pharmaceutically acceptable carriers, excipients, or diluents.
  • pharmaceutically acceptable carriers are well known to those skilled in the art and can be prepared in accordance with acceptable pharmaceutical procedures, such as, for example, those described in Remington 's Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, PA (1985), the entire disclosure of which is incorporated by reference herein for all purposes.
  • pharmaceutically acceptable refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient.
  • pharmaceutically acceptable carriers are those that are compatible with the other ingredients in the formulation and are biologically acceptable. Supplementary active ingredients can also be incorporated into the pharmaceutical compositions.
  • Compounds of the present teachings can be administered orally or parenterally, neat or in combination with conventional pharmaceutical carriers.
  • Applicable solid carriers can include one or more substances which can also act as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aids, binders or tablet-disintegrating agents, or encapsulating materials.
  • the compounds can be formulated in conventional manner.
  • Oral formulations containing a compound disclosed herein can comprise any conventionally used oral form, including tablets, capsules, buccal forms, troches, lozenges and oral liquids, suspensions or solutions.
  • the carrier in powders, can be a finely divided solid, which is an admixture with a finely divided compound.
  • a compound disclosed herein can be mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets can contain up to 99 % of the compound.
  • Capsules can contain mixtures of one or more compound(s) disclosed herein with inert filler(s) and/or diluent(s) such as pharmaceutically acceptable starches (e.g., corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses (e.g., crystalline and microcrystalline celluloses), flours, gelatins, gums, and the like.
  • inert filler(s) and/or diluent(s) such as pharmaceutically acceptable starches (e.g., corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses (e.g., crystalline and microcrystalline celluloses), flours, gelatins, gums, and the like.
  • Useful tablet formulations can be made by conventional compression, wet granulation or dry granulation methods and utilize pharmaceutically acceptable diluents, binding agents, lubricants, disintegrants, surface modifying agents (including surfactants), suspending or stabilizing agents, including, but not limited to, magnesium stearate, stearic acid, sodium lauryl sulfate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, carboxymethylcellulose calcium, polyvinylpyrrolidine, alginic acid, acacia gum, xanthan gum, sodium citrate, complex silicates, calcium carbonate, glycine, sucrose, sorbitol, dicalcium phosphate, calcium sulfate, lactose, kaolin, mannitol, sodium chloride, low melting waxes, and ion exchange resins.
  • pharmaceutically acceptable diluents including
  • Surface modifying agents include nonionic and anionic surface modifying agents.
  • Representative examples of surface modifying agents include, but are not limited to, poloxamer 188, benzalkonium chloride, calcium stearate, cetostearl alcohol, cetomacrogol emulsifying wax, sorbitan esters, colloidal silicon dioxide, phosphates, sodium dodecylsulfate, magnesium aluminum silicate, and triethanolamine.
  • Oral formulations herein can utilize standard delay or time-release formulations to alter the absorption of the compound(s).
  • the oral formulation can also consist of administering a compound disclosed herein in water or fruit juice, containing appropriate solubilizers or emulsifiers as needed.
  • Liquid carriers can be used in preparing solutions, suspensions, emulsions, syrups, elixirs, and for inhaled delivery.
  • a compound of the present teachings can be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, or a mixture of both, or a pharmaceutically acceptable oils or fats.
  • the liquid carrier can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers, and osmo-regulators.
  • liquid carriers for oral and parenteral administration include, but are not limited to, water (particularly containing additives as described herein, e.g., cellulose derivatives such as a sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) and their derivatives, and oils (e.g., fractionated coconut oil and arachis oil).
  • the carrier can be an oily ester such as ethyl oleate and isopropyl myristate.
  • Sterile liquid carriers are used in sterile liquid form compositions for parenteral administration.
  • the liquid carrier for pressurized compositions can be halogenated hydrocarbon or other pharmaceutically acceptable propellants.
  • Liquid pharmaceutical compositions which are sterile solutions or suspensions, can be utilized by, for example, intramuscular, intraperitoneal or subcutaneous injection. Sterile solutions can also be administered intravenously.
  • Compositions for oral administration can be in either liquid or solid form.
  • the pharmaceutical composition is in unit dosage form, for example, as tablets, capsules, powders, solutions, suspensions, emulsions, granules, or suppositories.
  • the pharmaceutical composition can be sub-divided in unit dose(s) containing appropriate quantities of the compound.
  • the unit dosage forms can be packaged compositions, for example, packeted powders, vials, ampoules, prefilled syringes or sachets containing liquids.
  • the unit dosage form can be a capsule or tablet itself, or it can be the appropriate number of any such compositions in package form.
  • Such unit dosage form can contain from about 1 mg/kg of compound to about 500 mg/kg of compound, and can be given in a single dose or in two or more doses.
  • Such doses can be administered in any manner useful in directing the compound(s) to the recipient's bloodstream, including orally, via implants, parenterally (including intravenous, intraperitoneal and subcutaneous injections), rectally, vaginally, and transdermally.
  • parenterally including intravenous, intraperitoneal and subcutaneous injections
  • rectally rectally, vaginally, and transdermally.
  • transdermally transdermally.
  • a compound of the present teachings can be provided to a patient already suffering from a disease in an amount sufficient to cure or at least partially ameliorate the symptoms of the disease and its complications.
  • the dosage to be used in the treatment of a specific individual typically must be subjectively determined by the attending physician.
  • the variables involved include the specific condition and its state as well as the size, age and response pattern of the patient.
  • the compounds of the present teachings can be formulated into a liquid composition, a solid composition, or an aerosol composition.
  • the liquid composition can include, by way of illustration, one or more compounds of the present teachings dissolved, partially dissolved, or suspended in one or more pharmaceutically acceptable solvents and can be administered by, for example, a pump or a squeeze-actuated nebulized spray dispenser.
  • the solvents can be, for example, isotonic saline or bacteriostatic water.
  • the solid composition can be, by way of illustration, a powder preparation including one or more compounds of the present teachings intermixed with lactose or other inert powders that are acceptable for intrabronchial use, and can be administered by, for example, an aerosol dispenser or a device that breaks or punctures a capsule encasing the solid composition and delivers the solid composition for inhalation.
  • the aerosol composition can include, by way of illustration, one or more compounds of the present teachings, propellants, surfactants, and co-solvents, and can be administered by, for example, a metered device.
  • the propellants can be a chlorofluorocarbon (CFC), a hydrofluoroalkane (HFA), or other propellants that are physiologically and environmentally acceptable.
  • compositions described herein can be administered parenterally or intraperitoneally. Solutions or suspensions of these compounds or a pharmaceutically acceptable salts, hydrates, or esters thereof can be prepared in water suitably mixed with a surfactant such as hydroxyl-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations typically contain a preservative to inhibit the growth of microorganisms.
  • the pharmaceutical forms suitable for injection can include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form can sterile and its viscosity permits it to flow through a syringe.
  • the form preferably is stable under the conditions of manufacture and storage and can be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • Compounds described herein can be administered transdermally, i.e., administered across the surface of the body and the inner linings of bodily passages including epithelial and mucosal tissues. Such administration can be carried out using the compounds of the present teachings including pharmaceutically acceptable salts, hydrates, or esters thereof, in lotions, creams, foams, patches, suspensions, solutions, and suppositories (rectal and vaginal).
  • Transdermal administration can be accomplished through the use of a transdermal patch containing a compound, such as a compound disclosed herein, and a carrier that can be inert to the compound, can be non-toxic to the skin, and can allow delivery of the compound for systemic absorption into the blood stream via the skin.
  • the carrier can take any number of forms such as creams and ointments, pastes, gels, and occlusive devices.
  • the creams and ointments can be viscous liquid or semisolid emulsions of either the oil-in-water or water-in-oil type. Pastes comprised of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the compound can also be suitable.
  • occlusive devices can be used to release the compound into the blood stream, such as a semi-permeable membrane covering a reservoir containing the compound with or without a carrier, or a matrix containing the compound.
  • Other occlusive devices are known in the literature.
  • Compounds described herein can be administered rectally or vaginally in the form of a conventional suppository.
  • Suppository formulations can be made from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerin.
  • Water-soluble suppository bases such as polyethylene glycols of various molecular weights, can also be used.
  • Lipid formulations or nanocapsules can be used to introduce compounds of the present teachings into host cells either in vitro or in vivo. Lipid formulations and nanocapsules can be prepared by methods known in the art.
  • the compounds of embodiments described herein can be administered in the conventional manner by any route where they are active. Administration can be systemic, topical, or oral. For example, administration can be, but is not limited to, parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, oral, buccal, or ocular routes, or intravaginally, by inhalation, by depot injections, or by implants.
  • modes of administration for the compounds of embodiments described herein can be, but are not limited to, sublingual, injectable (including short-acting, depot, implant and pellet forms injected subcutaneous ly or intramuscularly), or by use of vaginal creams, suppositories, pessaries, vaginal rings, rectal suppositories, intrauterine devices, and transdermal forms such as patches and creams.
  • Specific modes of administration will depend on the indication.
  • the selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response.
  • the amount of compound to be administered is that amount which is therapeutically effective.
  • the dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular animal treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician).
  • compositions containing the compounds of embodiments described herein and a suitable carrier can be solid dosage forms which include, but are not limited to, tablets, capsules, cachets, pellets, pills, powders and granules; topical dosage forms which include, but are not limited to, solutions, powders, fluid emulsions, fluid suspensions, semi-solids, ointments, pastes, creams, gels and jellies, and foams; and parenteral dosage forms which include, but are not limited to, solutions, suspensions, emulsions, and dry powder; comprising an effective amount of a polymer or copolymer of embodiments described herein.
  • the active ingredients can be contained in such formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • pharmaceutically acceptable diluents fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • the means and methods for administration are known in the art and an artisan can refer to various pharmacologic references for guidance. For example, Modern Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Gilman's The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980) can be consulted
  • the compounds of embodiments described herein can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • the compounds can be administered by continuous infusion subcutaneously over a period of about 15 minutes to about 24 hours.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the compounds can be formulated readily by combining these compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP).
  • disintegrating agents can be added, such as, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores can be provided with suitable coatings.
  • suitable coatings can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions can take the form of, e.g., tablets or lozenges formulated in a conventional manner.
  • the compounds for use according to embodiments described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon
  • compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds of embodiments described herein can also be formulated as a depot preparation.
  • Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • suitable polymeric or hydrophobic materials for example as an emulsion in an acceptable oil
  • ion exchange resins for example, ion exchange resins
  • sparingly soluble derivatives for example, as a sparingly soluble salt.
  • the compounds of embodiments described herein for example, can be applied to a plaster, or can be applied by transdermal, therapeutic systems that are consequently supplied to the organism.
  • compositions of the compounds also can comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as, e.g., polyethylene glycols.
  • compositions of embodiments described herein can also be administered in combination with other active ingredients, such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • active ingredients such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • the disintegrant component comprises one or more of croscarmellose sodium, carmellose calcium, crospovidone, alginic acid, sodium alginate, potassium alginate, calcium alginate, an ion exchange resin, an effervescent system based on food acids and an alkaline carbonate component, clay, talc, starch, pregelatinized starch, sodium starch glycolate, cellulose floe, carboxymethylcellulose, hydroxypropylcellulose, calcium silicate, a metal carbonate, sodium bicarbonate, calcium citrate, or calcium phosphate.
  • the diluent component comprises one or more of mannitol, lactose, sucrose, maltodextrin, sorbitol, xylitol, powdered cellulose, microcrystalline cellulose, carboxymethylcellulose, carboxyethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, methylhydroxyethylcellulose, starch, sodium starch glycolate, pregelatinized starch, a calcium phosphate, a metal carbonate, a metal oxide, or a metal aluminosilicate.
  • the optional lubricant component when present, comprises one or more of stearic acid, metallic stearate, sodium stearyl fumarate, fatty acid, fatty alcohol, fatty acid ester, glyceryl behenate, mineral oil, vegetable oil, paraffin, leucine, silica, silicic acid, talc, propylene glycol fatty acid ester, polyethoxylated castor oil, polyethylene glycol, polypropylene glycol, polyalkylene glycol, polyoxyethylene- glycerol fatty ester, polyoxyethylene fatty alcohol ether, polyethoxylated sterol, polyethoxylated castor oil, polyethoxylated vegetable oil, or sodium chloride.
  • a compound can be combined with other agents effective in the treatment of the target disease.
  • other active compounds i.e., other active ingredients or agents
  • the other agents can be administered at the same time or at different times than the compounds disclosed herein.
  • Compounds of the present teachings can be useful for the treatment or inhibition of a pathological condition or disorder in a mammal, for example, a human subject.
  • the present teachings accordingly provide methods of treating or inhibiting a pathological condition or disorder by providing to a mammal a compound of the present teachings inclding its pharmaceutically acceptable salt) or a pharmaceutical composition that includes one or more compounds of the present teachings in combination or association with pharmaceutically acceptable carriers.
  • Compounds of the present teachings can be administered alone or in combination with other therapeutically effective compounds or therapies for the treatment or inhibition of the pathological condition or disorder.
  • compositions according to embodiments described herein include from about 0.001 mg to about 1000 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 0.01 mg to about 100 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 100 mg to about 250 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 250 mg to about 500 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 500 mg to about 750 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 750 mg to about 1000 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; and from about 0.1 mg to about 10 mg of one or more compounds of the disclosure according to embodiments described herein; and one or more excipients.
  • compositions according to embodiments described herein are administered orally to a patient once daily.
  • compositions according to embodiments described herein are administered orally to a patient twice daily.
  • compositions according to embodiments described herein are administered orally to a patient three time per day.
  • compositions according to embodiments described herein are administered orally to a patient once weekly.
  • Cypl7 assay protocol AD293 cells that stably over-express recombinant CYP-17 were seeded in 96 well plates coated with poly D-lysine (15,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 ⁇ , Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS Fetal Bovine Serum
  • Cyp21 assay protocol AD293 cells that stably over-express recombinant CYP-21 were seeded in 96 well plates coated with poly D-lysine (10,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 ⁇ , Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS Fetal Bovine Serum
  • Cypll assay protocol AD293 cells that stably over-express recombinant CYP- 11 were seeded in 96 well plates coated with poly D-lysine (15,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS Fetal Bovine Serum
  • Table 23 Representative examples of compounds of the disclosure and their potencies in Cypl7, Cypl 1, and Cyp21 assays.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Diabetes (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Cardiology (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Psychiatry (AREA)
  • Emergency Medicine (AREA)
  • Vascular Medicine (AREA)
  • Child & Adolescent Psychology (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Pharmaceutical compositions described in this document comprise 5-(phenoxymethyl)-1,3-dioxane analogs having a disease-modifying action in the treatment of diseases associated with the production of cortisol that include metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, cancer, stroke, incidentalomas, or any diseases involving the overproduction of cortisol.

Description

NOVEL FUNC TIONALIZED 5-(PHENOXYMETHYL)-l,3-DIOXANE ANALOGS EXHIBITNG CYTOCHROME P450 INHIBITION
BRIEF SUMMARY
[0001] Embodiments described in this document are directed toward novel compounds of the formula (I),
Figure imgf000002_0001
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, prodrugs and complexes thereof, wherein:
[0002] Q is selected from a group consisting of optionally substituted aryl, optionally substituted
Figure imgf000002_0002
heteroaryl,
[0003] Rla, Rlb, Rlc, Rld, and Rle are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, C1-6, optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -
CO2R6, -C02NR4aR4b, -NHSO2R7, -SH, -SR7, S02R7and -SO2NHR6;
[0004] R2a, R2b, R2c, R2d, R2e, R2f and R2g are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6 optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -
C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7 and -S02NHR6;
[0005] R3 is selected from a group consisting of hydrogen, -S02R8, -C(0)NR9R10,
Figure imgf000002_0003
C(0)R7 -C(0)OR7, NH2j ^, and [0006] R a and R are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0007] R5 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0008] R6 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0009] R7 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0010] R8 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted C e haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl;
[0011] R9 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7
cycloalkyl, optionally substituted Ci_6 hal
Figure imgf000003_0001
[0012] R is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted C e branched alkyl;
[0013] Rl la and Rl lb are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH2OR6, and CE Heteroaryl.
[0014] Some embodiments described herein relate to a composition comprising an effective amount of at least one compounds according to the embodiments described in this document and at least one excipient.
[0015] Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve overproduction of Cortisol, including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the disease that involves overproduction of Cortisol is treated, delayed, slowed, or inhibited.
[0016] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve overproduction of Cortisol, including, for example, metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0017] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases or conditions associated with metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, or diseases that involve overproduction of Cortisol. Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
[0018] Some embodiments relate to a method of modulating Cortisol activity, the method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the compound or composition modulates Cortisol. In some embodiments, the compound or composition lowers Cortisol levels in the subject.
[0019] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas and diseases that involve overproduction of Cortisol, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient. [0020] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with overproduction of Cortisol. Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
[0021] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of disease or conditions associated with overproduction of Cortisol, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0022] Some embodiments yet further relate to a method of lowering the concentration of Cortisol in the circulatory system. Said methods comprise administering to a subject an effective amount of a compound or composition according to embodiments described herein.
[0023] Some embodiments yet further relate to a method of lowering the concentration of Cortisol in the circulatory system, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0024] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl7 activity, including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the disease that involves excess Cypl7 activity is treated, delayed, slowed, or inhibited.
[0025] Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl7 activity, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to the embodiments described herein and an excipient.
[0026] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl7 activity, including, for example, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered, and wherein the disease that is associated with Cypl7 activity is treated, delayed, slowed, or inhibited.
[0027] Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl7 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein Cypl7 activity is lowered.
[0028] Some embodiments also relate to a method for lowering Cypl7 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered. In some embodiments, lowering of Cypl7 activity leads to a lowering of testosterone levels to castrate levels in the subject. In some embodiments, lowering of Cypl7 activity leads to a lowering of estrogen levels to post- menopausal levels in the subject. Some embodiments are directed to a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments described herein, wherein Cypl7 activity is lowered. In some embodiments, Cypl7 activity is inhibited almost completely or completely. In some embodiments, lowering of Cypl7 activity leads to a lowering of testosterone levels to castrate levels in the subject. In some embodiments, lowering of Cypl7 activity leads to a lowering of estrogen levels to post- menopausal levels in the subject.
[0029] Some embodiments relate to a method of lowering Cypl7 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0030] Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl lBl activity, including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the disease that involves excess Cypl lB l activity is treated, delayed, slowed, or inhibited.
[0031] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cypl lBl activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0032] Some embodiments relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl lBl activity, including, for example, prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the Cypl lB activity is lowered and wherein the disease that involves excess Cypl 1B1 activity is treated, delayed, slowed, or inhibited.
[0033] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cypl lBl activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein the Cypl 1B1 activity is lowered.
[0034] Some embodiments also relate to a method for lowering Cypl lBl activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl 7 activity is lowered. Some embodiments relate to a method of lowering Cy l 1B1 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0035] Some embodiments relate to a method of inhibiting Cypl lBl activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0036] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the disease that involves excess Cyp21 activity is treated, delayed, slowed, or inhibited.
[0037] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases that involve excess Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, wherein said method comprises administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0038] Some embodiments also relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein Cyp21 activity is lowered, and wherein the disease that is associated with Cyp21 activity is treated, delayed, slowed, or inhibited.
[0039] Some embodiments yet further relate to a method for treating, delaying, slowing, or inhibiting the progression of diseases associated with Cyp21 activity, including, for example, androgenic hormones and estrogens are involved, such as prostate cancer, prostatic hypertrophy (prostatism), androgenic syndrome (masculinization), andromorphous baldness, breast cancer, mastopathy, uterine cancer, hirsutism, uterine fibroids, PCOS (polycystic ovarian syndrome), endometriosis, and ovarian cancer, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient, wherein the Cyp21 activity is lowered.
[0040] Some embodiments also relate to a method for lowering Cyp21 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments, wherein the Cypl7 activity is lowered. Some embodiments relate to a method of lowering Cyp21 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0041] Some embodiments relate to a method of inhibiting Cyp21 activity, said method comprising administering to a subject a composition comprising an effective amount of one or more compounds according to embodiments described herein and an excipient.
[0042] Some embodiments also relate to a method for lowering at least two of the following: Cypl7 activity, Cypl lBl activity, and Cyp21 activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or composition according to embodiments described herein. In some embodiments, the method further modulates Cortisol. Some embodiments relate to a method of treating, delaying, slowing, or inhibiting the progression of a disease selected from metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, related conditions, or a combination thereof, the method comprising administering to a subject in need thereof an effective amount of a compound or composition according to embodiments described herein, wherein the compound or composition lowers at least two of the following: Cypl7 activity, Cypl lBl activity, and Cyp21 activity in the subject. In some embodiments, the compound or composition modulates Cortisol. In some embodiments, the compound or composition lowers Cypl7 activity, Cypl lBl activity, and Cyp21 activity in the subject.
[0043] Some embodiments further relate to a process for preparing the compounds of embodiments described herein.
[0044] These and other objects, features, and advantages will become apparent to those of ordinary skill in the art from a reading of the following detailed description and the appended claims. All percentages, ratios and proportions herein are by weight, unless otherwise specified. All temperatures are in degrees Celsius (°C) unless otherwise specified. All documents cited are in relevant part, incorporated herein by reference; the citation of any document is not to be construed as an admission that it is prior art with respect to embodiments described herein.
DETAILED DESCRIPTION
[0045] Embodiments of the present invention describe novel compounds useful for the treatment of diseases associated with the production of Cortisol, such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, incidentalomas, and related conditions. In some embodiments, the diseases that involve production of Cortisol comprise diseases that involve an overproduction of Cortisol. In some embodiments, diabetes mellitus includes diabetes mellitus type I, diabetes mellitus type II, prediabetes, latent autoimmune diabetes of adults (LAD A), congenital diabetes, cystic fibrosis-related diabetes, steroid diabetes, monogenic diabetes, gestational diabetes, or a combination thereof.
[0046] Cortisol is a principal human glucocorticoid exhibiting many important physiological functions. It is involved in the regulation of the metabolism of proteins, carbohydrates, and fats; it counteracts insulin, maintains blood pressure and cardiovascular function, and suppresses the immune system's inflammatory response. However, pathological changes in adrenal and the upstream regulating switches can cause an overproduction of Cortisol. One disease associated with overproduction of Cortisol is metabolic syndrome. Over the course of the last three decades, a growing body of knowledge has been developed to describe metabolic syndrome, also referred to as "Syndrome X" or "Insulin Resistance Syndrome" (Reaven, G. M. Role of insulin resistance in human disease, Diabetes, 1988, 37, 1595-1607). Metabolic syndrome is defined as a cluster of abnormalities that occur in concert, including high blood pressure (BP), hyperglycemia, reduced high density lipoprotein cholesterol (HDL-C) levels, elevated triglycerides (TG) and abdominal obesity. The most widely accepted definition of this condition is based on the National Cholesterol Education Program (NCEP) Adult Treatment Panel-ΙΠ (ATP-III), which provides for the diagnosis of metabolic syndrome in patients that meet at least three of parameters identified in table 1. Current estimates indicate that nearly 25% of the world's adult population suffers from metabolic syndrome, and the incidence is rising, largely as a result of increased obesity rates (Anagnostis, P.; Athyros, V. G.; Tziomalos, K.; Karagiannis, A.; Dimitri P. Mikhailidis, D. P. The Pathogenetic role of Cortisol in the Metabolic Syndrome: A hypothesis, J. Clin. Endocrinol. Metab. 2009 94, 8, 2692-2701.).
Table 1 : Metabolic Syndrome diagnostic parameters
[0047] Cortisol production is regulated by several factors, including the enzymatic activity of the Ι ΐ β-hydroxylase (Cypl lBl), 17a-hydroxylase-C17,20-lyase (Cypl7), and 21 -hydroxylase (Cyp21). All three are members of the cytochrome P450 superfamily of enzymes. The 17a-hydroxylase/Ci7_2o lyase enzyme complex is essential for the biosynthesis of androgens. CYP17 is a bifunctional enzyme which possess both a C17-2o- lyase activity and a C17-hydroxylase activity. These two alternative enzymatic activities of CYP17 result in the formation of critically different intermediates in steroid biosynthesis and each activity appear to be differentially and developmentally regulated.
[0048] Cypl lB l catalyzes the final step of Cortisol synthesis, hydroxylation of the C-l l position of deoxycortisol. Cypl7 has multiple functions in corticosteroid synthesis. The C-17 and C-20 positions of the steroid framework can be modified by this enzyme. Pregnenolone and progesterone are hydroxylated by Cypl7 at C-17 (hydroxylase activity), while the C-20/C-17 bond is cleaved by the same enzyme in 17-hydroxyprogesterone and 17-hydroxypregnenolone (lyase activity). Finally, Cyp21 catalyzes the hydroxylation of C-21 in steroids such as progesterone and 17a-hydroxy progesterone.
[0049] Compounds that inhibit the enzymatic activity of Cypl7, Cyp21, or Cypl lBl will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas. Further, compounds that are dual inhibitors of Cypl7 and Cyp21 will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas. In addition, compounds that are dual inhibitors of Cypl7 and Cypl lBl will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas. Further, compounds that are dual inhibitors of Cypl lBl and Cyp21 will lead to a decrease in the synthesis of Cortisol, which would treat, delay, slow, or inhibit the progression of diseases associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
[0050] There is a long felt need for new treatments for diseases and symptoms associated with the overproduction of Cortisol such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas , that are both disease- modifying and effective in treating patients. Embodiments of the present invention addresses the need to identify effective treatment for diseases and symptoms associated with the overproduction of Cortisol, such as metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
[0051] The Cortisol lowering agents of embodiments described herein are capable of treating, delaying, slowing, or inhibiting the progression of diseases associated with the overproduction of Cortisol such as, for example, metabolic syndrome. It has been discovered that Cortisol is a principal human glucocorticoid exhibiting many important physiological functions. It is involved in the regulation of the metabolism of proteins, carbohydrates, and fats; it counteracts insulin, maintains blood pressure and cardiovascular function, and suppresses the immune system's inflammatory response. However, pathological changes in adrenal gland or other tissues capable of secreting Cortisol and the upstream regulating switches can cause an overproduction of Cortisol. One disease associated with overproduction of Cortisol is metabolic syndrome. In addition, the overproduction of Cortisol is associated with hypertension, diabetes mellitus, obesity, headache, depression, Cushing's syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas. Without wishing to be limited by theory, it is believed that Cortisol lowering agents of embodiments described in this disclosure ameliorate, abate, otherwise cause to be controlled, diseases associated with the overproduction of Cortisol, for example metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke and incidentalomas.
[0052] Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes are described as having, including, or comprising specific process steps, it is contemplated that compositions of the present teachings also consist essentially of, or consist of, the recited components, and that the processes of the present teachings also consist essentially of, or consist of, the recited processing steps.
[0053] As used herein, the term "consists of or "consisting of means that the method, use of formulation includes only the elements, steps, or ingredients specifically recited in the particular claimed embodiment or claim.
[0054] As used herein, the term "consisting essentially of or "consists essentially of means that the only active pharmaceutical ingredient in the formulation or method that treats the specified condition (e.g. Cushing's syndrome) is the specifically recited active pharmaceutical ingredient for treating the specified condition in the particular embodiment or claim; that is, the scope of the claim or embodiment is limited to the specified elements or steps and those that do not materially affect the basic and novel characteristic(s) of the particular embodiment or claimed invention.
[0055] In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components or a combination thereof, and can be selected from a group consisting of two or more of the recited elements or components.
[0056] The use of the singular herein includes the plural (and vice versa) unless specifically stated otherwise. In addition, where the use of the term "about" is before a quantitative value, the present teachings also include the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term "about" means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45%-55%.
[0057] It should be understood that the order of steps or order for performing certain actions is immaterial so long as the present teachings remain operable. Moreover, two or more steps or actions can be conducted simultaneously
[0058] As used herein, the term "excess" refers to an amount or quantity surpassing what is considered normal or sufficient. For example, excess Cypl7 activity may refer to an above normal level of the C17-hydroxylase activity of CYP17 which promotes the overproduction of glucocorticoids or an above normal level of the C17,20-lyase activity of Cypl7 which promotes the overproduction of sex hormones. In some embodiments, excess Cypl7 activity may lead to overproduction of Cortisol or an overproduction of androgenic or estrogenic hormones [0059] As used herein, the term "halogen" includes chlorine, bromine, fluorine, iodine, or a combination thereof.
[0060] As used herein, unless otherwise noted, "alkyl" and/or "aliphatic" whether used alone or as part of a substituent group refers to straight and branched carbon chains having 1 to 20 carbon atoms or any number within this range, for example 1 to 6 carbon atoms or 1 to 4 carbon atoms. Designated numbers of carbon atoms (e.g. C^) refers independently to the number of carbon atoms in an alkyl moiety or to the alkyl portion of a larger alkyl-containing substituent. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, z'so-propyl, n-butyl, sec-butyl, z'so-butyl, tert-butyl, and the like. Alkyl groups can be optionally substituted. Non-limiting examples of substituted alkyl groups include hydroxymethyl, chloromethyl, trifluoromethyl, aminomethyl, 1- chloroethyl, 2-hydroxyethyl, 1,2-difluoroethyl, 3-carboxypropyl, and the like. In substituent groups with multiple alkyl groups such as (Ci_6alkyl)2amino, the alkyl groups may be the same or different.
[0061] As used herein, the terms "alkenyl" and "alkynyl" groups, whether used alone or as part of a substituent group, refer to straight and branched carbon chains having 2 or more carbon atoms, preferably 2 to 20, wherein an alkenyl chain has at least one double bond in the chain and an alkynyl chain has at least one triple bond in the chain. Alkenyl and alkynyl groups can be optionally substituted. Non-limiting examples of alkenyl groups include ethenyl, 3-propenyl, 1-propenyl (also 2-methylethenyl), isopropenyl (also 2-methylethen-2-yl), buten-4-yl, and the like. Non-limiting examples of substituted alkenyl groups include 2-chloroethenyl (also 2-chlorovinyl), 4-hydroxybuten-l-yl, 7- hydroxy-7-methyloct-4-en-2-yl, 7-hydroxy-7-methyloct-3,5-dien-2-yl, and the like. Non- limiting examples of alkynyl groups include ethynyl, prop-2-ynyl (also propargyl), propyn-l-yl, and 2-methyl-hex-4-yn-l-yl. Non-limiting examples of substituted alkynyl groups include, 5-hydroxy-5-methylhex-3-ynyl, 6-hydroxy-6-methylhept-3-yn-2-yl, 5- hydroxy-5-ethylhept-3-ynyl, and the like.
[0062] As used herein, "cycloalkyl," whether used alone or as part of another group, refers to a non-aromatic carbon-containing ring including cyclized alkyl, alkenyl, and alkynyl groups, e.g., having from 3 to 14 ring carbon atoms, preferably from 3 to 7 or 3 to 6 ring carbon atoms, or even 3 to 4 ring carbon atoms, and optionally containing one or more (e.g., 1, 2, or 3) double or triple bond. In some embodiments, cycloalkyl groups may be monocyclic (e.g., cyclohexyl) or polycyclic (e.g., containing fused, bridged, and/or spiro ring systems), wherein the carbon atoms are located inside or outside of the ring system. Any suitable ring position of the cycloalkyl group can be covalently linked to the defined chemical structure. In some embodiments, cycloalkyl rings may be optionally substituted. Non-limiting examples of cycloalkyl groups include: cyclopropyl, 2-methyl-cyclopropyl, cyclopropenyl, cyclobutyl, 2,3-dihydroxycyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctanyl, decalinyl, 2,5-dimethylcyclopentyl, 3,5-dichlorocyclohexyl, 4- hydroxycyclohexyl, 3,3,5-trimethylcyclohex-l-yl, octahydropentalenyl, octahydro-lH- indenyl, 3a,4,5,6,7,7a-hexahydro-3H-inden-4-yl, decahydroazulenyl; bicyclo[6.2.0]decanyl, decahydronaphthalenyl, and dodecahydro-lH-fluorenyl. The term "cycloalkyl" also includes carbocyclic rings which are bicyclic hydrocarbon rings, non- limiting examples of which include, bicyclo-[2.1.1]hexanyl, bicyclo[2.2.1]heptanyl, bicyclo[3.1.1]heptanyl, l,3-dimethyl[2.2.1]heptan-2-yl, bicyclo[2.2.2]octanyl, and bicyclo [3.3.3 ]undecanyl.
[0063] As used herein, the term "haloalkyl" may include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogen. Haloalkyl groups include perhaloalkyl groups, wherein all hydrogens of an alkyl group have been replaced with halogens (e.g., -CF3, -CF2CF3). Haloalkyl groups can optionally be substituted with one or more substituents in addition to halogen. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, dichloroethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl groups.
[0064] As used herein, the term "alkoxy" refers to the group -O-alkyl, wherein the alkyl group is as defined above. Alkoxy groups optionally may be substituted. The term C3-C6 cyclic alkoxy refers to a ring containing 3 to 6 carbon atoms and at least one oxygen atom (e.g., tetrahydrofuran, tetrahydro-2H-pyran). C3-C6 cyclic alkoxy groups optionally may be substituted.
[0065] The term "aryl," wherein used alone or as part of another group, is defined herein as a an unsaturated, aromatic monocyclic ring of 6 carbon members or to an unsaturated, aromatic polycyclic ring of from 10 to 14 carbon members. Aryl rings can be, for example, phenyl or naphthyl ring each optionally substituted with one or more moieties capable of replacing one or more hydrogen atoms. Non-limiting examples of aryl groups include: phenyl, naphthylen-l-yl, naphthylen-2-yl, 4-fluorophenyl, 2-hydroxyphenyl, 3- methylphenyl, 2-amino-4-fluorophenyl, 2-(N,N-diethylamino)phenyl, 2-cyanophenyl, 2,6- di-tert-butylphenyl, 3-methoxyphenyl, 8-hydroxynaphthylen-2-yl 4,5- dimethoxynaphthylen-l-yl, and 6-cyano-naphthylen-l-yl. Aryl groups also include, for example, phenyl or naphthyl rings fused with one or more saturated or partially saturated carbon rings (e.g., bicyclo[4.2.0]octa-l,3,5-trienyl, indanyl), which can be substituted at one or more carbon atoms of the aromatic and/or saturated or partially saturated rings.
[0066] As used herein , the term "arylalkyl" or "aralkyl" refers to the group -alkyl-aryl, where the alkyl and aryl groups are as defined herein. Aralkyl groups of embodiments described herein are optionally substituted. Examples of arylalkyl groups include, for example, benzyl, 1 -phenylethyl, 2-phenylethyl, 3-phenylpropyl, 2-phenylpropyl, fluorenylmethyl and the like.
[0067] The terms "heterocyclic" and/or "heterocycle" and/or "heterocylyl," whether used alone or as part of another group, are defined herein as one or more ring having from 3 to 20 atoms wherein at least one atom in at least one ring is a heteroatom selected from nitrogen (Ν), oxygen (O), or sulfur (S), and wherein further the ring that includes the heteroatom is non-aromatic. In heterocycle groups that include 2 or more fused rings, the non-heteroatom bearing ring may be aryl (e.g., indolinyl, tetrahydroquinolinyl, chromanyl). Exemplary heterocycle groups have from 3 to 14 ring atoms of which from 1 to 5 are heteroatoms independently selected from nitrogen (Ν), oxygen (O), or sulfur (S). One or more Ν or S atoms in a heterocycle group can be oxidized. Heterocycle groups can be optionally substituted.
[0068] Non-limiting examples of heterocyclic units having a single ring include: diazirinyl, aziridinyl, urazolyl, azetidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazolidinyl, isothiazolyl, isothiazolinyl oxathiazolidinonyl, oxazolidinonyl, hydantoinyl, tetrahydrofuranyl, pyrrolidinyl, morpholinyl, piperazinyl, piperidinyl, dihydropyranyl, tetrahydropyranyl, piperidin-2-onyl (valerolactam), 2,3,4,5- tetrahydro-lH-azepinyl, 2,3-dihydro-lH-indole, and 1,2,3,4-tetrahydro-quinoline. Non- limiting examples of heterocyclic units having 2 or more rings include: hexahydro-lH- pyrrolizinyl, 3a,4,5,6,7,7a-hexahydro-lH-benzo[d]imidazolyl, 3a,4,5,6,7,7a-hexahydro- lH-indolyl, 1,2,3,4-tetrahydroquinolinyl, chromanyl, isochromanyl, indolinyl, isoindolinyl, and decahydro-lH-cycloocta[b]pyrrolyl.
[0069] The term "heteroaryl," whether used alone or as part of another group, is defined herein as one or more rings having from 5 to 20 atoms wherein at least one atom in at least one ring is a heteroatom chosen from nitrogen (N), oxygen (O), or sulfur (S), and wherein further at least one of the rings that includes a heteroatom is aromatic. In heteroaryl groups that include 2 or more fused rings, the non-heteroatom bearing ring may be a carbocycle (e.g., 6,7-Dihydro-5H-cyclopentapyrimidine) or aryl (e.g., benzofuranyl, benzothiophenyl, indolyl). Exemplary heteroaryl groups have from 5 to 14 ring atoms and contain from 1 to 5 ring heteroatoms independently selected from nitrogen (N), oxygen (O), or sulfur (S). One or more N or S atoms in a heteroaryl group can be oxidized. Heteroaryl groups can be substituted. Non-limiting examples of heteroaryl rings containing a single ring include: 1,2,3,4-tetrazolyl, [l,2,3]triazolyl, [l,2,4]triazolyl, triazinyl, thiazolyl, lH-imidazolyl, oxazolyl, furanyl, thiopheneyl, pyrimidinyl, 2- phenylpyrimidinyl, pyridinyl, 3-methylpyridinyl, and 4-dimethylaminopyridinyl. Non- limiting examples of heteroaryl rings containing 2 or more fused rings include: benzofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, cinnolinyl, naphthyridinyl, phenanthridinyl, 7H-purinyl, 9H-purinyl, 6-amino-9H-purinyl, 5H- pyrrolo[3,2-( Jpyrimidinyl, 7H-pyrrolo[2,3-( Jpyrimidinyl, pyrido[2,3-<i]pyrimidinyl, 2- phenylbenzo[d]thiazolyl, lH-indolyl, 4,5,6,7-tetrahydro-l-H-indolyl, quinoxalinyl, 5- methylquinoxalinyl, quinazolinyl, quinolinyl, 8-hydroxy-quinolinyl, and isoquinolinyl.
[0070] One non-limiting example of a heteroaryl group as described above is C1-C5 heteroaryl, which has 1 to 5 carbon ring atoms and at least one additional ring atom that is a heteroatom (preferably 1 to 4 additional ring atoms that are heteroatoms) independently selected from nitrogen (N), oxygen (O), or sulfur (S). Examples of C1-C5 heteroaryl include, but are not limited to, triazinyl, thiazol-2-yl, thiazol-4-yl, imidazol-l-yl, 1H- imidazol-2-yl, lH-imidazol-4-yl, isoxazolin-5-yl, furan-2-yl, furan-3-yl, thiophen-2-yl, thiophen-4-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridin-2-yl, pyridin-3-yl, and pyridin-4-yl.
[0071] Unless otherwise noted, when two substituents are taken together to form a ring having a specified number of ring atoms (e.g., R2 and R3 taken together with the nitrogen (N) to which they are attached to form a ring having from 3 to 7 ring members), the ring can have carbon atoms and optionally one or more (e.g., 1 to 3) additional heteroatoms independently selected from nitrogen (N), oxygen (O), or sulfur (S). The ring can be saturated or partially saturated and can be optionally substituted.
[0072] For the purposes of embodiments described herein fused ring units, as well as spirocyclic rings, bicyclic rings and the like, which comprise a single heteroatom will be considered to belong to the cyclic family corresponding to the heteroatom containing ring. For example, 1,2,3,4-tetrahydro ing the formula:
Figure imgf000019_0001
is, for the purposes of embodiments described herein, considered a heterocyclic unit. 6,7- Dihydro-5H-cyclopentapyrimidine hav formula:
Figure imgf000019_0002
is, for the purposes of embodiments described herein, considered a heteroaryl unit. When a fused ring unit contains heteroatoms in both a saturated and an aryl ring, the aryl ring will predominate and determine the type of category to which the ring is assigned. For example, l,2,3,4-tetrahydro-[l ,8]naphthyridine having the formula:
Figure imgf000019_0003
is, for the purposes of embodiments described herein, considered a heteroaryl unit.
[0073] Whenever a term or either of their prefix roots appear in a name of a substituent the name is to be interpreted as including those limitations provided herein. For example, whenever the term "alkyl" or "aryl" or either of their prefix roots appear in a name of a substituent (e.g., arylalkyl, alkylamino) the name is to be interpreted as including those limitations given above for "alkyl" and "aryl."
[0074] The term "substituted" is used throughout the specification. The term "substituted" is defined herein as a moiety, whether acyclic or cyclic, which has one or more hydrogen atoms replaced by a substituent or several (e.g., 1 to 10) substituents as defined herein below. The substituents are capable of replacing one or two hydrogen atoms of a single moiety at a time. In addition, these substituents can replace two hydrogen atoms on two adjacent carbons to form said substituent, new moiety or unit. For example, a substituted unit that requires a single hydrogen atom replacement includes halogen, hydroxyl, and the like. A two hydrogen atom replacement includes carbonyl, oximino, and the like. A two hydrogen atom replacement from adjacent carbon atoms includes epoxy, and the like. The term "substituted" is used throughout the present specification to indicate that a moiety can have one or more of the hydrogen atoms replaced by a substituent. When a moiety is described as "substituted" any number of the hydrogen atoms may be replaced. For example, difluoromethyl is a substituted Ci alkyl; trifluoromethyl is a substituted Ci alkyl; 4-hydroxyphenyl is a substituted aromatic ring; (N,N-dimethyl-5-amino)octanyl is a substituted Cs alkyl; 3-guanidinopropyl is a substituted C3 alkyl; and 2-carboxypyridinyl is a substituted heteroaryl.
[0075] The variable groups defined herein, e.g., alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryloxy, aryl, heterocycle and heteroaryl groups defined herein, whether used alone or as part of another group, can be optionally substituted. Optionally substituted groups will be so indicated.
[0076] The following are non-limiting examples of substituents which can substitute for hydrogen atoms on a moiety: halogen (chlorine (CI), bromine (Br), fluorine (F) and iodine®), -CN, -N02, oxo (=0), -OR12, -SR12, -N(R12)2, -NR12C(0)R12, -S02R12, - S02OR12, -S02N(R12)2, -C(0)R12, -C(0)OR12, -C(0)N(R12)2, Ci_6 alkyl, Ci_6 haloalkyl, Ci-6 alkoxy, C2_s alkenyl, C2_s alkynyl, C3-14 cycloalkyl, aryl, heterocycle, or heteroaryl, wherein each of the alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, aryl, heterocycle, and heteroaryl groups is optionally substituted with 1-10 (e.g., 1-6 or 1-4) groups selected independently from halogen, -CN, -N02, oxo, and R12; wherein R12, at each occurrence, independently is hydrogen, -OR13, -SR13, -C(0)R13, -C(0)OR13, - C(0)N(R13)2, -S02R13, -S(0)2OR13, -N(R13)2, -NR13C(0)R13, Ci_6 alkyl, Ci_6 haloalkyl, C2-8 alkenyl, C2-8 alkynyl, cycloalkyl (e.g., C3-6 cycloalkyl), aryl, heterocycle, or heteroaryl, or two R12 units taken together with the atom(s) to which they are bound form an optionally substituted carbocycle or heterocycle wherein said carbocycle or heterocycle has 3 to 7 ring atoms; wherein R13, at each occurrence, independently is hydrogen, Ci_6 alkyl, Ci_6 haloalkyl, C2_s alkenyl, C2_s alkynyl, cycloalkyl (e.g., C3-6 cycloalkyl), aryl, heterocycle, or heteroaryl, or two R13 units taken together with the atom(s) to which they are bound form an optionally substituted carbocycle or heterocycle wherein said carbocycle or heterocycle preferably has 3 to 7 ring atoms.
[0077] In some embodiments, the substituents are selected from
i) -OR14; for example, -OH, -OCH3, -OCH2CH3, -OCH2CH2CH3;
ii) -C(0)R14; for example, -COCH3, -COCH2CH3, -COCH2CH2CH3;
iii) -C(0)OR14; for example, -C02CH3, -C02CH2CH3, -C02CH2CH2CH3; iv) -C(0)N(R14)2; for example, -CONH2, -CONHCH3, -CON(CH3)2;
v) -N(R14)2; for example, -NH2, -NHCH3, -N(CH3)2, -NH(CH2CH3);
vi) halogen: -F, -CI, -Br, and -I; vii) -CHeXg; wherein X is halogen, m is from 0 to 2, e+g =3; for example,
-CH2F, -CHF2, -CF3, -CCI3, or -CBr3;
viii) -SO2R14; for example, -S02H; -S02CH:
ix) C1-C6 linear, branched, or cyclic alkyl;
x) Cyano
xi) Nitro;
xii) N(R14)C(0)R14;
xiii) Oxo (=0);
xiv) Heterocycle; and
xv) Heteroaryl.
wherein each R is independently hydrogen, optionally substituted C1-C6 linear or branched alkyl (e.g., optionally substituted C1-C4 linear or branched alkyl), or optionally substituted C3-C6 cycloalkyl (e.g optionally substituted C3-C4 cycloalkyl); or two R14 units can be taken together to form a ring comprising 3-7 ring atoms. In certain aspects, each R14 is independently hydrogen, C1-C6 linear or branched alkyl optionally substituted with halogen or C3-C6 cycloalkyl or C3-C6 cycloalkyl.
[0078] At various places in the present specification, substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges. For example, the term "Ci_6 alkyl" is specifically intended to individually disclose Ci, C2, C3, C4, C5, Ce, Ci-Ce, C1-C5, C -C4, C -C3, C1-C2, C2-C6, C2-C5, C2-C4, C2-C3, C3-C6, C3-C5, C3-C4, C4-C6, C4-C5, and C5-C6, alkyl.
[0079] For the purposes of embodiments described herein the terms "compound," "analog," and "composition of matter" stand equally well for the Cortisol lowering agent described herein, including all enantiomeric forms, diastereomeric forms, salts, and the like, and the terms "compound," "analog," and "composition of matter" are used interchangeably throughout the present specification.
[0080] Compounds described herein can contain an asymmetric atom (also referred as a chiral center), and some of the compounds can contain one or more asymmetric atoms or centers, which can thus give rise to optical isomers (enantiomers) and diastereomers. The present teachings and compounds disclosed herein include such enantiomers and diastereomers, as well as the racemic and resolved, enantiomerically pure R and S stereoisomers, as well as other mixtures of the R and S stereoisomers and pharmaceutically acceptable salts thereof. Optical isomers can be obtained in pure form by standard procedures known to those skilled in the art, which include, but are not limited to, diastereomeric salt formation, kinetic resolution, and asymmetric synthesis. The present teachings also encompass cis and trans isomers of compounds containing alkenyl moieties (e.g., alkenes and imines). It is also understood that the present teachings encompass all possible regioisomers, and mixtures thereof, which can be obtained in pure form by standard separation procedures known to those skilled in the art, and include, but are not limited to, column chromatography, thin-layer chromatography, and high-performance liquid chromatography.
[0081] Pharmaceutically acceptable salts of compounds of the present teachings, which can have an acidic moiety, can be formed using organic and inorganic bases. Both mono and polyanionic salts are contemplated, depending on the number of acidic hydrogens available for deprotonation. Suitable salts formed with bases include metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium, or magnesium salts; ammonia salts and organic amine salts, such as those formed with morpholine, thiomorpholine, piperidine, pyrrolidine, a mono-, di- or tri-lower alkylamine (e.g., ethyl- tert-butyl-, diethyl-, diisopropyl-, triethyl-, tributyl- or dimethylpropylamine), or a mono-, di-, or trihydroxy lower alkylamine (e.g., mono-, di- or triethanolamine). Specific non- limiting examples of inorganic bases include aHC03, a2C03, KHCO3, K2CO3, CS2CO3, LiOH, NaOH, KOH, NaH2P04, Na2HP04, and Na3P04. Internal salts also can be formed. Similarly, when a compound disclosed herein contains a basic moiety, salts can be formed using organic and inorganic acids. For example, salts can be formed from the following acids: acetic, propionic, lactic, benzenesulfonic, benzoic, camphorsulfonic, citric, tartaric, succinic, dichloroacetic, ethenesulfonic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, malonic, mandelic, methanesulfonic, mucic, napthalenesulfonic, nitric, oxalic, pamoic, pantothenic, phosphoric, phthalic, propionic, succinic, sulfuric, tartaric, toluenesulfonic, and camphorsulfonic as well as other known pharmaceutically acceptable acids.
[0082] When any variable occurs more than one time in any constituent or in any formula, its definition in each occurrence is independent of its definition at every other occurrence (e.g., in N(R13)2, each R13 may be the same or different than the other). Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. [0083] The terms "treat" and "treating" and "treatment" as used herein, refer to partially or completely alleviating, inhibiting, ameliorating and/or relieving a condition from which a patient is suspected to suffer.
[0084] As used herein, "therapeutically effective" and "effective dose" refer to a substance or an amount that elicits a desirable biological activity or effect.
[0085] A "therapeutically effective amount" or "effective amount" of a composition is a predetermined amount calculated to achieve the desired effect, i.e. treat, delay, slow, or inhibit the progression of diseases that involve overproduction of Cortisol. The activity contemplated by the present methods includes both medical therapeutic and/or prophylactic treatment, as appropriate. The specific dose of a compound administered according to this invention to obtain therapeutic and/or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the compound administered, the route of administration, and the condition being treated. The compounds are effective over a wide dosage range and, for example, dosages per day will normally fall within the range of from 0.001 to 10 mg/kg, more usually in the range of from 0.01 to 1 mg/kg. However, it will be understood that the effective amount administered will be determined by the physician in the light of the relevant circumstances including the condition to be treated, the choice of compound to be administered, and the chosen route of administration, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. A therapeutically effective amount of compound of this invention is typically an amount such that when it is administered in a physiologically tolerable excipient composition, it is sufficient to achieve an effective systemic concentration or local concentration in the tissue.
[0086] Except when noted, the terms "subject" or "patient" are used interchangeably and refer to mammals such as human patients and non-human primates, as well as experimental animals such as rabbits, rats, and mice, and other animals. Accordingly, the term "subject" or "patient" as used herein means any mammalian patient or subject to which the compounds of the invention can be administered. In an exemplary embodiment, to identify subject patients for treatment according to the methods of the invention, accepted screening methods are employed to determine risk factors associated with a targeted or suspected disease or condition or to determine the status of an existing disease or condition in a subject. These screening methods include, for example, conventional work-ups to determine risk factors that may be associated with the targeted or suspected disease or condition. These and other routine methods allow the clinician to select patients in need of therapy using the methods and compounds of embodiments described herein.
[0087] Embodiments described herein is directed toward novel compounds of the formula (I),
Figure imgf000024_0001
Including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, prodrugs and complexes thereof, wherein:
[0088] Q is selected from a group consisting of optionally substituted aryl, optionally substituted heteroaryl,
Figure imgf000024_0002
[0089] Rla, Rlb, Rlc, Rld, and Rle are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, C1-6, optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -
C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7and -S02NHR6;
[0090] R2a, R2b, R2c, R2d, R2e, R2f and R2g are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6 optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -
C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7 and -S02NHR6;
[0091] R3 is selected from a group consisting of hydrogen, -S02R8, -C(0)NR9R10, -
Figure imgf000024_0003
C(0)R7, -C(0)OR t7 , NH2j , and
[0092] R4a and R4b are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl; [0093] R5 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0094] R6 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0095] R7 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, and optionally substituted C3-7 cycloalkyl;
[0096] R8 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted C e haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl;
[0097] R9 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7
cycloalkyl, optionally substituted Ci_6 hal
Figure imgf000025_0001
[0098] R is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted C e branched alkyl;
[0099] Rl la and Rl lb are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH2OR6, and CH2Heteroaryl.
[0100] Some embodiments include com ounds having formula (II):
Figure imgf000025_0002
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0101] Some embodiments include compounds having formula (III):
Figure imgf000026_0001
including hydrates, solvates, enantiomers, diastereomers pharmaceutically acceptable salts, and complexes thereof.
[0102] Some embodiments include com ounds having formula (IV):
Figure imgf000026_0002
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0103] Some embodiments include compounds having formula (V):
Figure imgf000026_0003
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0104] Some embodiments include compounds having formula (VI):
Figure imgf000026_0004
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0105] Some embodiments include compounds having formula (VII):
Figure imgf000027_0001
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0106] Some embodiments include compounds having formula (VIII):
Figure imgf000027_0002
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0107] The embodiments of the present invention include compounds having formula (IX):
Figure imgf000027_0003
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0108] Some embodiments include compounds having formula (X):
Figure imgf000028_0001
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0109] Some embodiments include compounds having formula (Xa):
Figure imgf000028_0002
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, and complexes thereof.
[0110] In some embodiments Q is optionally substituted aryl.
[0111] In some embodiments Q is optionally substituted heteroaryl.
[0112] In some embodiments Q is
Figure imgf000028_0003
-fV
[0113] In some embodiments Q is \— / .
[0114] In some embodiments Rla of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7 and - S02NHR6.
[0115] In some embodiments Rlb of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0116] In some embodiments Rlc of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0117] In some embodiments Rld of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci-6 haloalkyl, Ci-6, optionally substituted alkoxy, - NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0118] In some embodiments Rle of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, - NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0119] In some embodiments R2a of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0120] In some embodiments R2b of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6. [0121] In some embodiments R2c of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0122] In some embodiments R2d of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0123] In some embodiments R2e of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0124] In some embodiments R2f of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6.
[0125] In some embodiments R2g of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_ 6 linear alkyl, optionally substituted Ci-6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, R2a is -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, -S02R7, and - S02NHR6. [0126] In some embodiments R3 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, -S02R8, -C(0)NR9R10, -C(0)R7, -
C(
Figure imgf000031_0001
[0127] In some embodiments R4a of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
[0128] In some embodiments R4b of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci-6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
[0129] In some embodiments R5 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alky, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
[0130] In some embodiments R6 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci-6 branched alkyl, and optionally substituted C3-7 cycloalkyl.
[0131] In some embodiments R7 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl.
[0132] In some embodiments R8 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl.
[0133] In some embodiments R9 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci-6 haloalkyl,
Figure imgf000031_0002
[0134] In some embodiments R10 of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted Ci-6 branched alkyl. [0135] In some embodiments Rlla of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH2OR6, and CH2Heteroaryl.
[0136] In some embodiments Rl lb of Formula I, II, III, IV, V, VI, VII, VIII, IX, X and Xa is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted aryl, optionally substituted benzyl, -CH2OR6, and CH2Heteroaryl.
[0137] Exemplary embodiments include compounds having the formula (X) or a pharmaceutically acceptable salt form thereof:
Figure imgf000032_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle, and R3 are defined herein below in Table 2.
Table 2:
Figure imgf000032_0002
Figure imgf000033_0001
[0138] Exemplary embodiments include compounds having the formula (XII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000034_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle, and R3 are defined herein below in Table 3.
Table 3:
Figure imgf000034_0002
Figure imgf000035_0001
[0139] Exemplary embodiments include compounds having the formula (XIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000035_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle, R9, and R10 are defined herein below in Table 4. Table 4:
Figure imgf000036_0002
[0140] Exemplary embodiments include compounds having the formula (XIV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000036_0001
wherein non-limiting examples of Rla, Rlb, Rlc, Rld, Rle, R9, and R10 are defined herein below in Table 5.
Table 5:
Figure imgf000036_0003
Entry R R b R R" R R R
5 CI H CI H H CH3 CH3
6 CI H CI H H CH2CH3 CH2CH3
7 CI H CI H H CH(CH3)2 CH(CH3)2
8 CI H CI H H cyclopropyl cyclopropyl
9 CI H H H H H CH3
10 CI H H H H H CH2CH3
11 CI H H H H H CH(CH3)2
12 CI H H H H H cyclopropyl
13 CI H H H H CH3 CH3
14 CI H H H H CH2CH3 CH2CH3
15 CI H H H H CH(CH3)2 CH(CH3)2
16 CI H H H H cyclopropyl cyclopropyl
[0141] Exemplary embodiments include compounds having the formula (XV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000037_0001
wherein non-limiting examples of Rla, Rlb, Rlc, Rld, Rle, Rl la, Rl lb, and R7 are defined herein below in Table 6.
Table 6:
Figure imgf000037_0002
Figure imgf000038_0001
[0142] Exemplary embodiments include compounds having the formula (XVI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000038_0002
wherein non-limiting examples of Rla, Rlb, Rlc, Rld, Rle, Rl la, Rl lb, and R7 are defined herein below in Table 7. Table 7:
Figure imgf000039_0001
[0143] Exemplary embodiments include compounds having the formula (XVII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000040_0001
wherein non-limiting examples of Rla, Rlb, Rlc, Rld, Rle, Rlla, Rllb, R4a, and R4b are defined herein below in Table 8.
Table 8:
Figure imgf000040_0002
Entry R R R R R ° R R 1 1 R R41
26 CI H H H H CH2Ph H CH2CH3 H
27 CI H H H H H CH2Ph CH3 H
28 CI H H H H H CH2Ph CH2CH3 H
[0144] Exemplary embodiments include compounds having the formula (XVIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000041_0001
wherein non-limiting examples of Rla, Rlb, Rlc, Rld, Rle, Rl la, Rl lb, R4a, and R4b are defined herein below in Table 9.
Table 9:
Figure imgf000041_0002
Entry R R R R R ° R R 1 1 R R41
19 CI H H H H H CH3 CH3 H
20 CI H H H H H CH3 CH2CH3 H
21 CI H H H H CH(CH3)2 H CH3 H
22 CI H H H H CH(CH3)2 H CH2CH3 H
23 CI H H H H H CH(CH3)2 CH3 H
24 CI H H H H H CH(CH3)2 CH2CH3 H
25 CI H H H H CH2Ph H CH3 H
26 CI H H H H CH2Ph H CH2CH3 H
27 CI H H H H H CH2Ph CH3 H
28 CI H H H H H CH2Ph CH2CH3 H
[0145] Exemplary embodiments include compounds having the formula (XIX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000042_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle, and R7 are defined herein below in Table 10.
Table 10:
Figure imgf000042_0002
[0146] Exemplary embodiments include compounds having the formula (XX) or a pharmaceutically acceptable salt form thereof:
Figure imgf000043_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle, and R7 are defined herein below in Table 1 1.
Table 11 :
Figure imgf000043_0003
[0147] Exemplary embodiments include compounds having the formula (XXI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000043_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in
Table 12. Table 12:
Figure imgf000044_0003
[0148] Exemplary embodiments include compounds having the formula (XXII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000044_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in
Table 13.
Table 13 :
Figure imgf000044_0004
[0149] Exemplary embodiments include compounds having the formula (XXIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000044_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in Table 14.
Table 14:
Figure imgf000044_0005
[0150] Exemplary embodiments include compounds having the formula (XXIV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000045_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in Table 15.
Table 15:
Figure imgf000045_0003
[0151] Exemplary embodiments include compounds having the formula (XXV) or a pharmaceutically acceptable salt form thereof:
Figure imgf000045_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in Table 16.
Table 16:
Figure imgf000045_0004
[0152] Exemplary embodiments include compounds having the formula (XXVI) or a pharmaceutically acceptable salt form thereof:
Figure imgf000046_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, and Rle are defined herein below in Table 17.
Table 17:
Figure imgf000046_0003
[0153] Exemplary embodiments include compounds having the formula (XXVII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000046_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle and R7 are defined herein below in Table 18.
Table 18:
Figure imgf000046_0004
[0154] Exemplary embodiments include compounds having the formula (XXVIII) or a pharmaceutically acceptable salt form thereof:
Figure imgf000047_0001
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle and R7 are defined herein below in Table 19.
Table 19:
Figure imgf000047_0004
[0155] Exemplary embodiments include compounds having the formula (XXVIIIa) or a pharmaceutically acceptable salt form thereof:
Figure imgf000047_0002
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle and R7 are defined herein below in Table 20.
Table 20:
Figure imgf000047_0005
[0156] Exemplary embodiments include compounds having the formula (XXVIIIb) or a pharmaceutically acceptable salt form thereof:
Figure imgf000047_0003
wherein non- limiting examples of Rla, Rlb, Rlc, Rld, Rle and R7 are defined herein below in Table 21.
Table 21 :
Figure imgf000048_0003
[0157] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXIX)
Figure imgf000048_0001
has the chemical name l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone.
[0158] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXX)
Figure imgf000048_0002
has the chemical name l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine.
[0159] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXI)
Figure imgf000049_0001
has the chemical name 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine-l- carboxamide.
[0160] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXII)
Figure imgf000049_0002
has the chemical name ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazine- 1 -carboxamido)acetate.
[0161] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXIII)
Figure imgf000049_0003
has the chemical name 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)- l,3-dioxan-5-yl)oxy)methyl)phenyl)-N-(2-(methylamino)-2-oxoethyl)piperazine-l- carboxamide.
[0162] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXIV) (XXXIV) has the chemical name methyl 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazine- 1 -carboxylate.
[0163] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXV)
Figure imgf000050_0001
has the chemical name 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)piperazine- 1 -carboximidamide.
[0164] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXVI)
Figure imgf000050_0002
has the chemical name 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)-4,5-dihydrooxazole.
[0165] For the purposes of demonstrating the manner in which the compounds of embodiments described herein are named and referred to herein, the compound having the formula (XXXVII)
Figure imgf000051_0001
has the chemical name 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)-5,6-dihydro-4H- 1,3- oxazine.
[0166]In this document, a compound depicted by the racemic formula, for example:
Figure imgf000051_0002
will stand equally well for either of the two enantiomers having the formula:
Figure imgf000051_0003
or the formula:
Figure imgf000051_0004
or mixtures thereof, or in the case where a third chiral center is present, all diastereomers.
[0167] In all of the embodiments provided herein, examples of suitable optional substituents are not intended to limit the scope of the claimed invention. The compounds of the invention may contain any of the substituents, or combinations of substituents, provided herein.
PROCESS [0168] Some embodiments of the present invention further relate to a process for preparing the Cortisol lowering agents of embodiments described herein.
[0169] Compounds of the present teachings can be prepared in accordance with the procedures outlined herein, from commercially available starting materials, compounds known in the literature, or readily prepared intermediates, by employing standard synthetic methods and procedures known to those skilled in the art. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be readily obtained from the relevant scientific literature or from standard textbooks in the field. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions can vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. Those skilled in the art of organic synthesis will recognize that the nature and order of the synthetic steps presented can be varied for the purpose of optimizing the formation of the compounds described herein.
[0170] The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., lH or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high pressure liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
[0171] Preparation of the compounds can involve protection and deprotection of various chemical groups. The need for protection and deprotection and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Greene et al, Protective Groups in Organic Synthesis, 2d. Ed. (Wiley & Sons, 1991), the entire disclosure of which is incorporated by reference herein for all purposes.
[0172] The reactions or the processes described herein can be carried out in suitable solvents which can be readily selected by one skilled in the art of organic synthesis. Suitable solvents typically are substantially nonreactive with the reactants, intermediates, and/or products at the temperatures at which the reactions are carried out, i.e., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected.
[0173] The compounds of these teachings can be prepared by methods known in the art of organic chemistry. The reagents used in the preparation of the compounds of these teachings can be either commercially obtained or can be prepared by standard procedures described in the literature. For example, compounds of embodiments described herein can be prepared according to the method illustrated in the General Synthetic Schemes.
GENERAL SYNTHETIC SCHEMES FOR PREPARATION OF COMPOUNDS.
[0174] The reagents used in the preparation of the compounds of this invention can be either commercially obtained or can be prepared by standard procedures described in the literature. In accordance with this invention, compounds in the genus may be produced by one of the following reaction schemes.
[0175] Compounds of the disclosure may be prepared according to the processes outlined in schemes 1-13.
Figure imgf000053_0001
[0176] A suitably substituted compound of formula (1), a known compound or compound prepared by known methods, is reacted with a bromine in an organic solvent such as 1 ,4- dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, N,N- dimethylformamide, and the like to provide a compound of the formula (2). A compound of the formula (2) is then reacted with trimethyl orthoacetate in the presence of an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like in a solvent such as methanol, 1,4-dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, and the like optionally with heating to provide a compound of the formula (3). A compound of the formula (3) is then reacted with a compound of the formula (4), a known compound or compound prepared by known methods, in the presence of an acid such as p-toluenesulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like in a solvent such as benzene, toluene, p- xylene, 1 ,4-dioxane, tetrahydrofuran, and the like to provide a compound of the formula (5). A compound of the formula (5) is then reacted with a compound of the formula (6), a known compound or compound prepared by known methods, in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like, in a solvent such as N,N-dimethylformamide, Ν,Ν-dimethylacetamide, 1,4-dioxane, tetrahydrofuran, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (7).
Figure imgf000054_0001
[0177] Benzaldehyde is reacted with a glycerol in an organic solvent such as 1,4-dioxane, tetrahydrofuran, 1,2-dichloroethane, Ν,Ν-dimethylformamide, N,N-dimethylacetamide, and the like, in the presence of an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (9). A compound of the formula (9) is then reacted with a compound of the formula (10) wherein X is a leaving group such as bromine, chlorine, methansulfonate, and the like, in the presence of a base such as sodium hydride, potassium hydride, lithium diisopropylamide, sodium diisopropylamide, lithium bis(trimethylsilyl)amide, sodium bis(trimethylsilyl)amide, and the like in an solvent such as 1,4-dioxane, tetrahydrofuran, ethyl ether, methylene chloride, 1,2-dichloroethane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (11). A compound of the formula (11) is then reacted with an acid such as p-toluenesulfonic acid, camphorsulfonic acid, hydrochloric acid, sulfuric acid, acetic acid, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (4).
Scheme 3
Figure imgf000055_0001
[0178] A compound of the formula (7) is reacted with a compound of the formula (12), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium, and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xylene, 1,4-dioxane, tetrahydrofuran, and the like, optionally in the presence of water, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (13).
Scheme 4
Figure imgf000055_0002
[0179] A compound of the formula (13) is reacted with a base such as a potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as methanol, ethanol, isopropanol, and the like, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (14). Scheme 5
[0180] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (15), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (16).
Figure imgf000056_0001
[0181] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (17), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (18).
Scheme 7
Figure imgf000056_0002
[0182] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (19), a known compound or compound prepared by known methods in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like to provide a compound of the formula (20).
Scheme 8
Figure imgf000057_0001
[0183] Alternatively, a suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a p- nitrophenylchloroformate in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (21). A compound of formula (21) is then reacted with a compound of the formula (22), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (20). Scheme 9
Figure imgf000058_0001
[0184] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (23), in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine,
2,6-dimethylpyridine, N-methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like to provide a compound of the formula (24).
Scheme 10
Figure imgf000058_0002
[0185] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (25), a known compound or compound prepared by known methods wherein n is 1 or 2, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1 ,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (26). A compound of formula (26) is then reacted with a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N-methylmorpholine, potassium carbonate, sodium carbonate, lithium carbonate, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, N,N- dimethylformamide, and the like to provide a compound of the formula (27). Scheme 1 1
Figure imgf000059_0001
[0186] A suitably substituted compound of formula (14), a known compound or compound prepared by known methods, is reacted with a compound of the formula (28), a known compound or compound prepared by known methods, in the presence of a bases such as such as triethylamine, diisopropylethylamine, pyridine, 2,6-dimethylpyridine, N- methylmorpholine, and the like, in an organic solvent such as methylene chloride, dichloroethane, tetrahydrofuran, 1,4-dioxane, Ν,Ν-dimethylformamide, and the like to provide a compound of the formula (29).
Figure imgf000059_0002
[0187] A compound of the formula (7) is reacted with a compound of the formula (30), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium, [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xylene, 1,4-dioxane, tetrahydrofuran, and the like, optionally in the presence of water, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (31).
Figure imgf000060_0001
[0188] A compound of the formula (7) is reacted with a compound of the formula (32), a known compound or compound prepared by known methods, in the presence of a palladium catalyst such as palladium acetate, palladium bis(triphenylphosphine) dichloride, palladium tetrakis(triphenylphospine), bis(acetonitrile)dichloropalladium, [l, l'-Bis(diphenylphosphino) ferrocene]dichloropalladium and the like, optionally in the presence of 2-Dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (X-phos), in the presence of a base such as potassium carbonate, sodium carbonate, lithium carbonate, cesium carbonate, sodium hydroxide, lithium hydroxide, potassium hydroxide, and the like in a solvent such as toluene, benzene, p-xylene, 1,4-dioxane, tetrahydrofuran, and the like, optionally in the presence of water, optionally with heating, optionally with microwave irradiation, to provide a compound of the formula (31).
[0189] The Examples provided below provide representative methods for preparing exemplary compounds of embodiments described herein. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds of embodiments described herein.
[0190] Examples 1-X provide methods for preparing representative compounds of the disclosure The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare additional compounds of embodiments described herein.
[0191] Example 1 : Synthesis of 2-Bromo-l-(2, 4-dichlorophenyl)ethanone.
Figure imgf000060_0002
[0192] To a stirred solution of l-(2,4-dichlorophenyl)ethanone (100 g, 0.528 mol) in 1,4- dioxane/ethyl ether (2: 1, 600 mL) bromine (27 mL, 0.528 mol) was added drop wise at 0 °C over 1 h and the reaction mixture was then stirred for 1 hour. The reaction mixture was poured into ice-water and extracted with ethyl acetate. The organic layer was washed with water, brine and dried ( a2S04) filtered and concentrated to give the title compound
(145 g, 98 %) as pale yellow liquid. The crude product was taken for the next step without any further purification. XH NMR (300 MHz, CDC1,) δ 7.56-7.52 (m, 1H),
7.48 (d, J= 1.8 Hz, 1H), 7.35 (dd, J= 1.8, 8.4 Hz, 1H), 4.49 (s, 2H).
[0193] The following compounds can be prepared by the procedure of 2-Bromo-l-(2, 4- dichlorophenyl) ethanone. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
[0194] Example 3 : Synthesis of 2-bromo-l-(2-chlorophenyl)ethanone:
Figure imgf000061_0001
[0195] The title compound was prepared according to the procedure for 2-Bromo-l-(2, 4- dichlorophenyl)ethanone, except l-(2-chlorophenyl)ethanone was substituted for l-(2,4- dichlorophenyl)ethanone. XH NMR (400 MHz, CDC13) δ 7.57 (d, J= 7.6 Hz, 1H), 7.45 (d, J= 3.6 Hz, 1H), 7.38-7.34 (m, 2H), 4.52 (s, 2H).
[0196] Example 4: Synthesis of l-(2-Bromo-l, l-dimethoxyethyl)-2,4-dichlorobenzene
Figure imgf000061_0002
[0197] To a stirred solution of 2-Bromo-l-(2,4-dichlorophenyl)ethanone (145 g, 0.541 mol) in dry methanol (500 mL) was added trimethyl orthoformate (172 mL, 1.623 mol), followed by toluene-4-sulfonic acid monohydrate (10.29 g, 0.054 mol) at room temperature and the reaction mixture was refluxed for 12 hours. Reaction mixture was quenched with sodium methoxide at 0 °C and extracted with ethyl acetate. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by column chromatography (100-200 mesh) eluting with 10% Ethyl acetate in petroleum ether to give the title compound. XH NMR: (400 MHz, CDC13) δ
7.79 (d, J = 8.4 Hz, 1H), 7.40 (d, J = 2.4 Hz, 1H), 7..29-7.26 (m, 1H), 3.89 (s, 2H), 3.22
(s, 6H). [0198] The following compounds can be prepared by the procedure of l-(2-Bromo-l, l- dimethoxyethyl)-2,4-dichlorobenzene. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
[0199] Example 5: Synthesis of l-(2-bromo-l, l-dimethoxyethyl)-2-chlorobenzene:
Figure imgf000062_0001
[0200] The title compound was prepared according to the procedure for l-(2-Bromo-l,l- dimethoxyethyl)-2,4-dichlorobenzene, except 2-bromo-l-(2-chlorophenyl)ethanone was substituted for 2-Bromo-l-(2,4-dichlorophenyl)ethanone. ¾ NMR: (300 MHz, CDC13) δ 7.87-7.82 (m, 1H), 7.38-7.35 (m, 1H), 7.29-7.25 (m, 2H), 3.93 (s, 2H), 3.22 (s, 6H).
[0201] Example 6: Synthesis of (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2- (2,4-dichlorophenyl)-l,3-dioxane and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)- 2-(2,4-dichlorophenyl)-l,3-dioxane.
Figure imgf000062_0002
[0202] A mixture of l-(2-Bromo-l, l-dimethoxyethyl)-2,4-dichlorobenzene (15 g, 0.0477 mol) and 2-(4-Bromobenzyloxy) propane-1, 3-diol (12.4 g, 0.0477 mol) in dry benzene (150 mL) was refluxed for 1 hour. After lhour solvent was distilled out (50 mL) and cooled to 60 °C. Toluene-4-sulfonic acid monohydrate was added at 60 °C to reaction mixture and refluxed for 3 hours. The reaction mixture was quenched with triethylamine, diluted with water and extracted with ethyl acetate. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated to give title compound as a mixture of isomers. The isomers were separated by column chromatography (100-200 mesh) using 10% ethyl acetate in petroleum ether. (2s,5s)-5-((4-bromobenzyl)oxy)-2- (bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane : ¾ NMR: (400 MHz, CDC13) δ 7.56 (d, J = 8.8 Hz, 1H), 7.48-7.46 (m, 3H), 7.35 (dd, J = 2.0, 8.4 Hz, 1H), 7.16 (d, J = 8.4 Hz, 2H), 4.48 (s, 2H), 4.15 (q, 2H), 3.9-3.83 (m, 1H), 3.61 (s, 2H), 3.44 (t, J = 10.4 Hz, 2H). (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3- dioxane: XH NMR: (400 MHz, CDC13) δ 7.55 (d, J = 8.8 Hz, 1H), 7.48-7.45 (m, 3H), 7.32-7.26 (m, 3H), 4.6 (s, 2H), 4.12 (d, J= 12.0 Hz, 2H), 3.78 (d, J = 12.4 Hz, 2H), 3.68 (s, 2H), 3.26 (t, J= 2.0 Hz, 1H).
[0203] The following compounds can be prepared by the procedure of (2s,5s)-5-((4- bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane and (2r,5r)-5- ((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
[0204] Example 7: Synthesis of (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)- 1 ,3 -dioxane.
[0205] The title compounds were prepared according to the procedure for (2s,5s)-5-((4- bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)-l,3-dioxane and (2r,5r)-5- ((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4-dichlorophenyl)- 1,3 -dioxane was substituted for 1 -(2-bromo- 1 , 1 -dimethoxyethyl)-2,4-dichlorobenzene.
Figure imgf000063_0001
[0206] (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2-chlorophi dioxane. 'H NMR: (300 MHz, CDC13) δ 7.60-7.55 (m, 1H), 7.45-7.39 (m, 3H), 7.37-7.27 (m, 2H), 7.13-7.10 (m, 2H), 4.46 (s, 2H), 4.14-4.09 (m, 2H), 3.94-3.82 (m, 1H), 3.62 (s, 2H), 3.49-3.42 (m, 2H).
Figure imgf000063_0002
[0207] (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane. XH NMR: (300 MHz, CDC13) δ 7.64-7.59 (m, 1H), 7.48-7.40 (m, 3H), 7.33-7.26 (m, 4H), 4.62 (s, 2H), 4.14-4.10 (m, 2H), 3.83-3.79 (m, 2H), 3.70 (s, 2H), 3.25-3.23 (m, 1H).
[0208] Example 8: Synthesis of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- 1 H-imidazole.
Figure imgf000064_0001
[0209] To a stirred solution of (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4- dichlorophenyl)-l,3-dioxane (l lg, 0.0215 mol) in dry N,N-dimethylformamide (120 mL) potassium carbonate (14.87 g, 0.107 mol) was added, followed by imidazole (14.65 g, 0.215 mol) at room temperature and the reaction mixture was refluxed for 48 hours. The reaction mixture was filtered to remove the precipitated solid and the filtrate was poured into ice-water and extracted with ethyl acetate. The organic layer was washed with water, brine and dried (Na2S04) filtered and concentrated and crude material was purified by column chromatography on silica (100-200 mesh) eluting with 50% ethyl acetate in petroleum ether to give the title compound. XH NMR: (400 MHz, CDC13) δ 7.45 (m, 3H), 7.27 (d, J = 5.6 Hz, 1H), 7.17 (s, 2H), 7.11 (d, J = 8.4 Hz, 2H), 6.9 (s, 1H), 6.71 (s, 1H), 4.42 (s, 2H), 4.17 (s, 2H), 4.03 (q, 2H), 3.72-3.64 (m, 1H), 3.36 (t, J= 9.6 Hz, 2H); LCMS: m/z = 498.19 [(M+H)+]; (Purity: 98%); IR (thin film): 2875.67, 1585.52, 1467.39, 1 107.06, 804.91 cm"1.
[0210] Example 9: Synthesis of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2,4- dichlorophenyl)-l,3-dioxa -2-yl)methyl)-lH-imidazole (7b):
Figure imgf000064_0002
[0211] To a stirred solution of (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4- dichlorophenyl)-l,3-dioxane (15 g, 0.0293 mol) in dry N,N-dimethylformamide (120 mL) potassium carbonate (20.2 g, 0.146 mol) was added, followed by imidazole (19.98 g, 0.293 mol) at room temperature and the reaction mixture was refluxed for 48 hours. The reaction mixture was filtered and the filtrate was poured into ice-water and extracted with ethyl acetate. The organic layer was washed with water, brine and dried ( a2S04) filtered and concentrated and the crude material was purified by column chromatography on silica (100-200 mesh) eluting with 50% ethyl acetate in petroleum ether to give the title compound. XH NMR: (400 MHz, CDC13) δ 7.5-7.45 (m, 3H), 7.24 (d, J = 4 Hz, 7.21 (s, 1H), 7.15 (dd, J= 2, 8, 8.0 Hz, 1H), 7.09 (d, J= 8.0 Hz, 1H), 6.87 (s, 1H) (s, 1H), 4.56 (s, 2H), 4.29 (s, 2H), 4.07 (d, J = 11.6 Hz, 2H), 3.74 (d, J = 1 1.6 2H), 3.21 (s, 1H); LCMS: m/z = 498. 19 [(M+H)+].
[0212] The following compounds can be prepared by the procedure of l-(((2s,5s)-5-((4- bromobenzyl)oxy)-2-(2,4-dichlorophenyl)-l,3-dioxan-2-yl)methyl)-lH-imidazole. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
[0213] Example 10: Synthesis of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- lH-imidazole.
Figure imgf000065_0001
[0214] The title compounds were prepared according to the procedure for l-(((2s,5s)-5- ((4-bromobenzyl)oxy)-2-(2,4-dichlorophenyl)-l,3-dioxan-2-yl)methyl)-lH-imidazole, except (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane was substituted for (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4- dichlorophenyl)-l,3-dioxane. 'H NMR: (400 MHz, CDC13) 7.45-7.42 (m, 3H), 7.30-7.27 (m, 2H), 7.25-7.24 (m, 1H), 7.22-7.17 (m, 1H), 7.11 (d, J = 8.4 Hz, 2H), 6.89 (s, 1H), 6.72 (s, 1H), 4.42 (s, 2H), 4.19 (s, 2H), 4.04-3.99 (m, 2H), 3.72-3.65 (m, 1H), 3.41-3.37 (m, 2H); ESIMS: m/z = 462.9 [(M+H)+].
[0215] Example 1 1 : Synthesis of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)- 1 ,3 -dioxan-2-yl)methyl)- lH-imidazole.
Figure imgf000065_0002
[0216] The title compounds were prepared according to the procedure for l-(((2s,5s)-5- ((4-bromobenzyl)oxy)-2-(2,4-dichlorophenyl)-l,3-dioxan-2-yl)methyl)-lH-imidazole, except (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane was substituted for (2s,5s)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2,4- dichlorophenyl)-l,3-dioxane. ¾ NMR: (400 MHz, CDC13) δ 7.49 (d, J = 8.4 Hz, 2H), 7.44 (d, J = 8.0 Hz, 1H), 7.29-7.27 (m, 1H), 7.23-7.21 (m, 3H), 7.17-7.15 (m, 2H), 6.85 (s, 1H), 6.70 (s, 1H), 4.56 (s, 2H), 4.30 (s, 2H), 4.09-4.05 (m, 2H), 3.80-3.75 (m, 2H), 3.21-3.20 (m, 1H); ESIMS: m/z = 462.9 [(M+H)+].
[0217] Example 12: Synthesis of 5-(4-Bromobenzyloxy)-2-phenyl-l, 3-dioxane
Figure imgf000066_0001
[0218] To a stirred solution of benzaldehyde (200 g, 1.88 mol) in dry N,N- dimethylformamide (500 mL) was added glycerol (344 mL, 4.71mol), followed by camphor- 10-sulphonic acid (43.8 g, 0.188 mol) at room temperature and the reaction mixture was heated to 70 °C for 2 hours. The reaction mixture was poured into ice-water and extracted with ethyl acetate and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine and dried (Na2S04) filtered and concentrated. The crude product was purified by column chromatography on silica (100-200 mesh) eluting with 15% ethyl acetate in petroleum ether to give an oil. The oil was dissolved in tetrahydrofuran (200 mL) and added dropwise over 45 minutes to a suspension of NaH (53.33 g, 2.22 mol) in tetrahydrofuran (800 mL). After 45 minutes 4- bromobenzylbromide (277.7 g, 1.1 1 mol) was added portionwise over 30 minutes and the reaction mixture was stirred at room temperature for 6 hours. The reaction mixture was poured into ice-water and extracted with ethyl acetate. The organic layer was washed with water, brine, dried ( a2S04) filtered and concentrated. The crude product was washed with petroleum ether and the precipitated solid was filtered. The solid product was purified by column chromatography on silica (100-200 mesh) eluting with 10% ethyl acetate in petroleumether to give the title compound. XH NMR: (300 MHz, CDC13) δ 7.49-7.44 (m, 4H), 7.37-7.34 (m, 3H), 7.21 (d, J = 8.4 Hz, 2H), 5.39 (s, 1H), 4.53 (s, 2H), 4.37 (q, 2H), 3.77-3.72 (m, 1H), 3.63 (t, J= 9.3 Hz, 2H); LCMS: m/z = 349.2 [(M+H)+].
[0219] Example 13: Synthesis of 2-(4-Bromobenzyloxy) propane-1, 3-diol
Figure imgf000066_0002
[0220] To a stirred solution of 5-(4-Bromobenzyloxy)-2 -phenyl- 1, 3-dioxane (20 g, 0.057 mol) in methanol (150 mL) dilute HC1 (0.5 N, 65 mL) was added dropwie at 0 °C over 15 minutes and the reaction mixture was refluxed for 30 minutes. The reaction mixture was poured into ice-water and washed with petroleum ether (2 x 150 mL). The aqueous layer was basified with 10% sodium bicarbonate solution and extracted with ethyl acetate and washed with water, brine, dried ( a2S04) filtered and concentrated to give the title compound. ¾ NMR: (300 MHz, DMSO-i¾ δ 7.53 (d, J= 8.4 Hz, 2H), 7.34 (d, J= 7.8 Hz, 2H), 4.58 (s, 2H), 4.56 (bs, 2H), 3.52-3.32 (m, 5H); IR (thin film): 3271.13, 1485.44, 1341.71, 1 120.78, 1070.56, 802.64 cm"1.
[0221] Example 14: Synthesis of l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone
Figure imgf000067_0001
[0222] To a stirred solution of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2,4- dichlorophenyl)-l,3-dioxan-2-yl)methyl)-lH-imidazole (4.5 g, 9.03 mmol) in dry toluene (70 mL) was added 1-acetylpiperazine (1.5 g, 1 1.74 mmol) followed by CS2CO3 (5.88 g, 18.06 mmol) and the reaction mixture was degassed with argon for 10 minutes. X-Phos (0.43 g, 0.903 mmol), palladium acetate (0.20 g, 0.903 mmol) were then added and again degassed with argon for 10 minutes. The reaction mixture was refluxed over 2 hours. The reaction mixture was filtered and concentrated. The crude product was purified by column chromatography on silica (100-200 mesh) eluting with 10% methanol in ethyl acetate to give the title compound. ¾-NMR: (400 MHz, CDC13) δ 7.44 (s, 1H), 7.29 (s, 1H), 7.16 (s, 1H), 7.15 (d, J = 8.0 Hz, 3H), 6.91 (s, 1H), 6.87 (d, J = 8.8 Hz, 2H), 6.71 (s, 1H), 4.39 (s, 2H), 4.17 (s, 2H), 4.0 (q, 2H), 3.76 (t, J = 5.6 Hz, 2H), 3.69-3.65 (m, 1H), 3.61 (t, J = 5.2 Hz, 2H), 3.36 (t, J = 9.2 Hz, 2H), 3.18-3.12 (m, 4H), 2.13 (s, 3H); LCMS: m/z = 545.45 [(M+H)+]. IR (thin film): 3465.45, 2923.08, 1643.94, 1436.44, 1233.85, 1078.20, 803.77 cm"1.
[0223] Example 15: Synthesis of l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone.
Figure imgf000068_0001
[0224] To a stirred solution of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2,4-dichlorophenyl)- l,3-dioxan-2-yl)methyl)-lH-imidazole (0.5 g, 1.00 mmol) in dry toluene (20 mL) was added 1-acetylpiperazine (0.16 g, 1.304 mmol) followed by CS2CO3 (0.65 g, 2.00 mmol) and the reaction mixture was degassed with argon for 10 minutes. X-Phos (0.047 g, 0.100 mmol), palladium acetate (0.022 g, 0.100 mmol) were added and again degassed with argon for 10 min. The reaction mixture was then refluxed for 12 hours. The reaction mixture was filtered and concentrated. The residue was purified by column chromatography on silica (100-200 mesh) eluting with 10% methanol in ethyl acetate to give the title compound. (400 MHz, CDC13) δ 7.45 (d, J = 2.0 Hz, 1H), 7.34 (s, 1H), 7.23 (d, J = 10.0 Hz, 2H), 7.12 (d, J = 2.0 Hz, 1H), 7.09 (d, J = 8.0 Hz, 1H), 6.92 (d, J = 8.8 Hz, 2H), 6.88 (s, 1H), 6.71 (s, 1H), 4.53 (s, 2H), 4.3 (s, 2H), 4.06 (d, J = 1 1.6 Hz, 2H), 3.78 (t, J = 5.6 Hz, 2H), 3.7 (t, J = 7.2 Hz, 2H), 3.63 (t, J = 5.6 Hz, 2H), 3.2 (s, 1H), 3.17 (m, 4H), 2.14 (s, 3H); LCMS: m/z = 545.47 [(M+H)+].
[0225] Example 16: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)piperazine.
Figure imgf000068_0002
[0226] To a stirred solution of l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)ethanone (0.6 g, 1.1 mmol) in methanol (5 mL) was added 30% NaOH (1 mL) and the reaction mixture was refluxed for 16 hours. The reaction mixture was poured into water and extracted with methylene chloride. The organic layer was washed with water, brine, dried ( a2S04) filtered and concentrated to give the title compound. ^- MR: (400 MHz, DMSOi&5) δ 7.66 (d, J = 1.6 Hz, 1H), 7.41-7.38 (dd, J = 1.6, 8 Hz, 1H), 7.25 (m, 1H), 7.09 (d, J = 9.2 Hz, 2H), 6.85 (d, J = 8.8 Hz, 2H), 6.8 (s, 1H), 6.74 (s, 1H), 4.35 (s, 2H), 4.26 (s, 2H), 4.05 (q, 2H), 3.63 (m, 1H), 3.19 (t, J = 20.4 Hz, 2H), 3 (t, J =10 Hz, 4H), 2.8 (t, J =9.2 Hz, 4H), 2.28 (bs, 1H). LCMS: m/z = 502.14 [(M+H)+].
[0227] Example 17: Synthesis of 3-((4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l, -dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)sulfonyl)benzonitrile.
Figure imgf000069_0001
[0228] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.05 g, 0.099 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.069 mL, 0.497 mmol) followed by 3-cyanobenzene sulfonyl chloride (0.024 g, 0.1 19 mmol) and the reaction mixture was stirred at room temperature over 1 hour. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound. ^- MR: (300 MHz, CDC13) δ 8.07 (s, 1H), 8.02 (d, J= 8.1 Hz, 1H), 7.91 (d, J= 7.5 Hz, 1H), 7.73-7.68 (m, 1H), 7.44 (s, 1H), 7.29 (s, 1H), 7.16-7.11 (m, 4H), 6.9 (s, 1H), 6.28 (d, J = 8.7 Hz, 2H), 6.71 (s, 1H), 4.37 (s, 2H), 4.16 (s, 2H), 4.0 (q, 2H), 3.67-3.62 (m, 1H), 3.36 (d, J = 9.9 Hz, 2H), 3.23-3.21 (m, 8H). LCMS: m/z = 670 [(M+H)+].
[0229] Example 18: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-((3 - chloropropyl)sulfonyl)piperazine.
Figure imgf000069_0002
[0230] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.08 g, 0.159 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.11 mL, 0.795 mmol) followed by 3-chloropropane sulfonyl chloride (0.023 mL, 0.190 mmol) and the reaction mixture was stirred at room temperature over lhour. The reaction mixture was poured into ice- water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound. ^- MR: (300 MHz, CDC13) δ 7.46 (s, 2H), 7.2 (s, 2H), 7.16 (d, J= 8.4 Hz, 2H), 6.97 (s, 1H), 6.88 (d, J= 8.7 Hz, 2H), 6.76 (s, 1H), 4.39 (s, 2H), 4.18 (s, 2H), 4.0 (q, 2H), 3.74-3.68 (m, 3H), 3.44 (t, J= 9.9 Hz, 4H), 3.38(d, J= 9.9 Hz, 2H ), 3.25 (t, J= 9.6 Hz, 4H), 3.12 (t, J= 14.7 Hz, 2H), 2.35-2.30 (m, 2H). LCMS: m/z = 643 [(M+H)+].
[0231] Example 19: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- lsulfonyl)piperazine.
Figure imgf000070_0001
[0232] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.08 g, 0.159 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.11 mL, 0.795 mmol) followed by cyclopropane sulfonyl chloride (0.026 g, 0.190 mmol) and the reaction mixture was stirred at room temperature over 2 hours. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound. ^- MR: (400 MHz, CDC13) δ 7.45 (s, 1H), 7.30 (s, 2H), 7.17-7.14 (m, 3H), 6.91 (s, 1H), 6.88 (d, J= 8.4 Hz, 2H), 6.71 (s, 1H), 4.39 (s, 2H), 4.17 (s, 2H), 4.01 (q, 2H), 3.69-3.65 (m, 1H), 3.45 (t, J= 10 Hz, 4H), 3.25 (t, J= 10 Hz, 4H). LCMS: m/z = 606.14 [(M+H)+]. [0233] Example 20: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- lfonyl)piperazine.
Figure imgf000071_0001
[0234] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.09 g, 0.178 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.12 mL, 0.894 mmol) followed by 2-propane sulfonyl chloride (0.03 g, 0.214 mmol) and the reaction mixture was stirred at room temperature over 1 hour. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried ( a2S04) filtered and concentrated. The crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound. ^- MR: (300 MHz, CDC13) δ 7.44 (s, 1H), 7.17-
7.13 (m, 5H), 6.91 (s, 1H), 6.88 (d, J = 8.4 Hz, 2H), 6.73 (s, 1H), 4.39 (s, 2H), 4.17 (s, 2H), 4.0 (q, 2H), 3.67-3.66 (m, 1H), 3.50 (t, J = 9.6 Hz, 4H), 3.37 (d, J= 10.2 Hz, 2H), 3.20-3.19 (m, 5H), 1.38 (s, 3H); 1.36(s, 3H). LCMS: m/z = 608.16 [(M+H)+].
[0235] Example 21 Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- l)piperazine.
Figure imgf000071_0002
To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.09 g, 0.178 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.12 mL, 0.894 mmol) followed by ethane sulfonyl chloride (0.027 g, 0.214 mmol) and the reaction mixture was stirred at room temperature over 1 hour. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried ( a2S04) filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound. ^- MR: (400 MHz, CDC13) δ 7.45 (s, 1H), 7.32 (s, 1H), 7.17-7.14 (m, 4H), 6.91 (s, 1H), 6.88 (d, J = 8.4 Hz, 2H), 6.72 (s, 1H), 4.39 (s, 2H), 4.17 (s, 2H), 4.01 (q, 2H), 3.67 (m, 1H), 3.44 (t, J = 10 Hz, 4H), 3.34 (s, 2H), 3.23 (t, J = 10 Hz, 4H), 3.02 (q, 2H), 1.40 (t, J = 15.2 Hz, 3H). LCMS: m/z = 594.14 [(M+H)+].
[0236] Example 22: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-(( 1 H-imidazol-4- yl)sulfonyl)piperazine.
Figure imgf000072_0001
[0237] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.09 g, 0.178 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.12 mL, 0.894 mmol) followed by lH-imidazole-4- sulfonyl chloride (0.035 g, 0.214 mmol) and the reaction mixture was stirred at room temperature over 1 hour. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound. ^- MR: (300 MHz, CDC13) δ 7.71 (s, 1H), 7.62(s, 1H), 7.45 (s, 2H), 7.19 (s, 2H), 7.12 (d, J= 8.4 Hz, 2H), 6.93(s, 1H), 6.82 (d, J= 8.7 Hz, 2H), 6.72 (s, 1H), 4.37 (s, 2H), 4.16 (s, 2H), 3.99 (q, 2H), 3.68-3.65 (m, 1H), 3.32 (s, 1H), (m, 8H), 3.23-3.22 (m, 8H). LCMS: m/z = 632.13 [(M+H)+].
[0238] Example 23: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)- 1 ,3 -dioxan-5 -yl)oxy)methyl)phenyl)-4-((3 - (trifluoromethoxy)phenyl)sulfonyl)piperazine.
Figure imgf000073_0001
[0239] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.09 g, 0.178 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.12 mL, 0.894 mmol) followed by 3-(trifluoromethoxy)benzene sulfonyl chloride (0.055 g, 0.214 mmol) and the reaction mixture was stirred at room temperature over 2 hours. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried (Na2S04) filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound. ^- MR: (400 MHz, CDC13) δ 7.73
(d, , J = 8 Hz, 1H), 7.64-7.59 (m, 2H), 7.48 (d, , J = 7.2 Hz 1H), 7.44 (s, 1H), 7.16 (s, 2H),7.13 (d, , J = 8 Hz, 2H), 6.9 (s, 1H), 6.82 (d, , J = 8 Hz 2H), 6.71 (s, 1H), 4.37 (s, 2H), 4.16 (s, 2H), 3.99 (q, 2H), 3.67-3.63 (m, 1H), 3.32 (s, 2H), 3.23-3.18 (m, 8H). LCMS: m/z = 726.12 [(M+H)+].
[0240] Example 24: Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3- sulfonyl)piperazine.
Figure imgf000073_0002
[0241] To a stirred solution of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine (0.08 g, 0.159 mmol) in dry methylene chloride (5 mL) was added triethylamine (0.1 1 mL, 0.795 mmol) followed by pyridine-3 -sulfonyl chloride (0.022 mL, 0.190 mmol) and the reaction mixture was stirred at room temperature over 1 hour. The reaction mixture was poured into ice-water and extracted with methylene chloride and washed with 10% sodium bicarbonate solution. The organic layer was washed with water, brine, dried ( a2S04) filtered and concentrated. The crude product was purified by prep TLC (10% methanol in methylene chloride) to give the title compound. ^- MR: (400 MHz, CDC13) 59.02 (d, , J = 2 Hz, 1H), 8.85-8.84 (m, 1H), 8.08 (d, , J= 8 Hz, 1H), 7.52-7.49 (m, 1H), 7.44 (s, 1H), 7.26 (s, 1H), 7.15-7.11 (m, 3H), 6.89 (s, 1H), 6.82 (d, , J = 8.4 Hz, 2H), 6.71 (s, 1H), 4.37 (s, 2H), 4.16 (s, 2H), 3.99 (q, 2H), 3.68-3.63 (m, 1H), 3.32 (s, 2H), 3.23-3.22 (m, 8H). LCMS: m/z = 643.14 [(M+H)+].
[0242] Example 26: Synthesis of l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)ethanone.
Figure imgf000074_0001
[0243] To a stirred solution of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)- l,3-dioxan-2-yl)methyl)-lH-imidazole (0.4 g, 0.87 mmol) in dry toluene (18 mL) was added 1-acetylpiperazine (0.13 g, 1.04 mmol) followed by CS2CO3 (0.56 g, 1.74 mmol) and the reaction mixture was degassed with argon for 10 minutes, X-Phos (0.041g, 0.09 mmol), palladium acetate (0.019 g, 0.09 mmol) were then added and again degassed with argon for 10 minutes. The reaction mixture was refluxed over 2 hours. The reaction mixture was filtered and concentrated and the residue was purified by column chromatography on silica (100-200 mesh) eluting with 10% methanol in ethyl acetate to afford the title compound. ^- MR: (400 MHz, CDC13) δ 7.43 (d, J= 7.6 Hz, 1H), 7.30- 7.26 (m, 2H), 7.25-7.23 (m, 1H), 7.20 (d, J = 6.8 Hz, 1H), 7.16 (d, J= 8.4 Hz, 2H), 6.89- 6.85 (m, 3H), 6.71 (s, 1H), 4.39 (s, 2H), 4.19 (s, 2H), 4.01-3.97 (m, 2H), 3.77-3.74 (m, 2H), 3.72-3.64 (m, 1H), 3.62-3.59 (m, 2H), 3.45-3.35 (m, 2H), 3.18-3.12 (m, 4H), 2.13 (s, 3H); ESIMS: m/z = 510.9 [(M+H)+].
[0244] Example 27: Synthesis of l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)ethanone.
Figure imgf000075_0001
To a stirred solution of l-(((2r,5r)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)-l,3- dioxan-2-yl)methyl)-lH-imidazole (0.4 g, 0.87 mmol) in dry toluene (18 mL) was added 1-acetylpiperazine (0.13 g, 1.04 mmol) followed by CS2CO3 (0.56 g, 1.74 mmol) and the reaction mixture was degassed with argon for 10 minutes. X-Phos (0.041 g, 0.09 mmol), palladium acetate (0.019 g, 0.09 mmol) were then added and again degassed with argon for 10 minutes. The reaction mixture was refluxed over 2 hours. The reaction mixture was filtered and concentrated. The crude product was purified by column chromatography on silica (100-200 mesh) eluting with 10% methanol in ethyl acetate to afford the title compound. XH NMR: (300 MHz, CDC13) δ 7.67-7.65 (m, 1H), 7.60-7.55 (m, 2H), 7.42-
7.38 (m, 2H), 7.30-7.28 (m, 1H), 7.26-7.22 (m, 3H), 6.92-6.89 (m, 2H), 4.48 (s, 2H), 4.41
(s, 2H), 4.07-4.02 (m, 2H), 3.94-3.90 (m, 1H), 3.80-3.75 (m, 4H), 3.64-3.62 (m, 2H),
3.20-3.14 (m, 4H), 2.14 (s, 3H).; ESIMS: m/z = 510.9 [(M+H)+].
[0245] Example 28: Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4- dichlorophenyl)-l,3-dio
Figure imgf000075_0002
To a stirred solution of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)-l,3- dioxan-2-yl)methyl)-lH-imidazole (0.3 g, 0.602 mmol) in 1 ,4-Dioxane/water (9: 1, lOmL) was added K2CO3 (0.24 g, 1.80 mmol), pyridine-4-boronicacid pinacol ester (0.148 g, 0.722 mmol) and the reaction mixture was degassed with nitrogen gas for 10 minutes. [l,l '-Bis(diphenylphosphino)ferrocene]dichloropalladium (0.044 g, 0.0602 mmol), was added and again degassed with nitrogen gas for 10 min.. The reaction mixture was then refluxed for 12 hours. The reaction mixture was filtered and concentrated. The crude product was purified by prep TLC (10% Methanol in methylene chloride) to give the title compound. XH-NMR: (400 MHz, CDC13) 58.65 (d, , J = 6 Hz, 2H), 7.6 (d, , J= 8.1 Hz, 2H), 7.48-7.46 (m, 3H), 7.36 (d, , J= 8.1 Hz, 2H), 7.29 (s, 1H), 7.29 (s, 1H), 7.18 (d, , J = 0.9 Hz, 2H), 6.9 (s, 1H), 6.73 (s, 1H), 4.53 (s, 2H), 4.18 (s, 2H), 4.07 (q, 2H), 3.76-3.7 (m, 1H), 3.39 (t, , J = 20.1 Hz, 2H). LCMS: m/z = 495.11 [(M+H)+].
[0246] Example 29: Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)morpholine.
Figure imgf000076_0001
To a stirred solution of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)-l,3- dioxan-2-yl)methyl)-lH-imidazole (0.28 g, 0.61 mmol) in dry toulene (5 mL) was added morpholine (0.063 g, 0.73 mmol) followed by CS2CO3 (0.39 g, 1.22 mmol) and the reaction mixture was degassed with argon for 10 minutes. X-Phos (0.028g, 0.06 mmol), palladium acetate (0.013 g, 0.06 mmol) were then added and again degassed with argon for 10 minutes. The reaction mixture was refluxed over 2 hours. The reaction mixture was filtered, concentrated and the residue was purified by prep TLC to afford the title compound. XH NMR: (300 MHz, CDC13) δ 7.43-7.40 (m, 1H), 7.26-7.13 (m, 6H), 6.90- 6.83 (m, 3H), 6.71 (s, 1H), 4.39 (s, 2H), 4.19 (s, 2H), 4.01-3.96 (m, 2H), 3.86-3.83 (m, 4H), 3.72-3.63 (m, 1H), 3.42-3.31 (m, 2H), 3.15-3.12 (m, 4H); ESIMS: m/z = 469.9
[(M+H)+].
[0247] Example 30: Synthesis of 4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)morpholine.
Figure imgf000076_0002
To a stirred solution of (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane (0.2 g, 0.43 mmol) in dry toulene (5 mL) was added morpholine (0.045 g, 0.5 mmol) followed by CS2CO3 (0.28 g, 0.86 mmol) and the reaction mixture was degassed with argon for 10 minutes. X-Phos (0.02 g, 0.04 mmol), palladium acetate (0.010 g, 0.04 mmol) were then added and again degassed with argon for 10 minutes. The reaction mixture was refluxed over 2 hours. The reaction mixture was filtered, concentrated and the residue was purified by prep TLC to afford the title compound. XH NMR: (400 MHz, CDC13) 57.43 (d, J = 8.0 Hz, 1H), 7.26-7.24 (m, 1H), 7.23-7.19 (m, 3H), 7.12-7.11 (m, 2H), 6.90 (d, J= 8.0 Hz, 2H), 6.83 (s, 1H), 6.66 (s, 1H), 4.56 (s, 2H), 4.32 (s, 2H), 4.06-4.03 (m, 2H), 3.87-3.84 (m, 4H), 3,76-3.73 (m, 2H), 3.18- 3.14 (m, 5H); ESIMS: m/z = 469.9 [(M+H)+].
[0248] The following compounds can be prepared by the procedure of 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine. The skilled practitioner will know how to substitute the appropriate reagents, starting materials and purification methods known to those skilled in the art, in order to prepare the compounds provided herein.
[0249] Example 31 : Synthesis of l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine:
[0250] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except 1 -(methylsulfonyl)piperazine was substituted for morpholine. XH NMR: (400 MHz, CDC13) δ 7.43 (d, J = 7.2 Hz, 1H), 7.29-7.26 (m, 2H), 7.25-7.24 (m, 1H), 7.20-7.14 (m, 3H), 6.90-6.86 (m, 6H), 6.72 (s, 1H), 4.39 (s, 2H), 4.19 (s, 2H), 4.02-3.97 (m, 2H), 3.71-3.64 (m, 1H), 3.38-3.36 (m, 6H), 3.28-3.25 (m, 4H), 2.82 (s, 3H); ESIMS: m/z = 547.03 [(M+H)+].
[0251] Example 32: Synthesis of l-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine:
Figure imgf000077_0002
[0252] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except 1 -(methylsulfonyl)piperazine was substituted for morpholine and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane was substituted for (2s,5s)-5-((4-bromobenzyl)oxy)-2- (bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane. XH NMR: (400 MHz, CDC13) δ 7.43 (d, J= 7.2 Hz, 1H), 7.29-7.26 (m, 2H), 7.25-7.24 (m, 1H), 7.20-7.14 (m, 3H), 6.90-6.86 (m, 6H), 6.72 (s, 1H), 4.39 (s, 2H), 4.19 (s, 2H), 4.02-3.97 (m, 2H), 3.71-3.64 (m, 1H), 3.38- 3.36 (m, 6H), 3.28-3.25 (m, 4H), 2.82 (s, 3H); ESIMS: m/z = 547.03 [(M+H)+].
[0253] Example 33 : ynthesis of 2-methoxy ethyl 4-(4-((((2s,5s)-2-((lH-imidazol-l- yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l- carboxylate:
Figure imgf000078_0001
[0254] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except 2-methoxy ethyl piperazine-l-carboxylate was substituted for morpholine. XH NMR: (400 MHz, CDC13) δ 7.43-7.41 (m, 1H), 7.31- 7.27 (m, 2H), 7.26-7.18 (m, 2H), 7.16-7.13 (m, 2H), 6.91 (s, 1H), 6.87 (d, J = 8.8 Hz, 2H), 6.71 (s, 1H), 4.38 (s, 2H), 4.28-4.25 (m, 2H), 4.19 (s, 2H), 4.01-3.97 (m, 2H), 3.71- 3.60 (m, 7H), 3.50-3.39 (m, 5H), 3.14-3.11 (m, 4H).; ESIMS: m/z = 571.1 [(M+H)+].
[0255] Example 34: Synthesis of 2-methoxyethyl 4-(4-((((2r,5r)-2-((lH-imidazol-l- yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l- carboxylate:
Figure imgf000078_0002
[0256] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except 2-methoxyethyl piperazine-l-carboxylate was substituted for morpholine and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane was substituted for (2s,5s)-5-((4-bromobenzyl)oxy)-2- (bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane. 'H NMR: (400 MHz, CDC13) δ 7.43 (d, J = 8.0 Hz, 1H), 7.24-7.20 (m, 3H), 7.14-7.11 (m, 3H), 6.91 (d, J = 8.4 Hz, 2H), 6.83 (s, 1H), 6.66 (s, 1H), 4.55 (s, 2H), 4.32 (s, 2H), 4.28-4.26 (m, 2H), 4.06-4.03 (m, 2H), 3.76- 3.73 (m, 2H), 3.65-3.61 (m, 6H), 3.39 (s, 3H), 3.18-3.14 (m, 5H); ESIMS: m/z = 571.3 [(M+H)+].
[0257] Example 35: Synthesis of ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2- (2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate:
Figure imgf000079_0001
[0258] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except ethyl 2-(piperazine-l-carboxamido)acetate was substituted for morpholine. XH NMR: (400 MHz, CDC13) δ 7.43 (d, J= 7.2 Hz, 1H), 7.28- 7.26 (m, 1H), 7.25-7.23 (m, 2H), 7.20 (d, J = 8.0 Hz, 1H), 7.15 (d, J= 8.4 Hz, 2H), 6.86- 6.84 (m, 3H), 6,75-6.65 (m, 1H), 5.02-4.98 (m, 1H), 4.39 (s, 2H), 4.25-4.19 (m, 4H), 4.03-3.97 (m, 4H), 3.69-3.65 (m, 1H), 3.56 (t, J= 5.2 Hz, 4H), 3.42-3.35 (m, 2H), 3.18 (t, J= 5.6 Hz, 4H), 1.31 (q, J= 6.8 Hz, 3H); ESIMS: m/z = 598.1 [(M+H)+].
[0259] Example 36: Synthesis of ethyl 2-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2- (2-chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate:
Figure imgf000079_0002
[0260] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except ethyl 2-(piperazine-l-carboxamido)acetate was substituted for morpholine and (2r,5r)-5-((4-bromobenzyl)oxy)-2-(bromomethyl)-2-(2- chlorophenyl)-l,3-dioxane was substituted for (2s,5s)-5-((4-bromobenzyl)oxy)-2- (bromomethyl)-2-(2-chlorophenyl)-l,3-dioxane. XH NMR: (400 MHz, CDC13) δ 7.43- 7.41 (m, 1H), 7.31-7.22 (m, 4H), 7.12-7.1 1 (m, 2H), 6.91-6.83 (m, 3H), 6.70-6.67 (m, 1H), 5.02-4.99 (m, 1H), 4.56 (s, 2H), 4.33 (s, 2H), 4.26-4.19 (m, 2H), 4.07-4.02 (m, 3H), 3.77-3.73 (m, 2H), 3.60-3.58 (m, 4H), 3.49 (s, 2H), 3.21-3.19 (m, 4H), 1.31 (q, J = 9.2 Hz, 3H).; ESIMS: m/z = 598.2 [(M+H)+].
[0261] Example 37: Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan-5-yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine- 1 - carboxamide.
Figure imgf000080_0001
[0262] The title compound was prepared according to the procedure for 4-(4-((((2s,5s)-2- ((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-5- yl)oxy)methyl)phenyl)morpholine, except Ν,Ν-dimethylpiperazine- 1 -carboxamide was substituted for morpholine. XH NMR: (400 MHz, CDC13) δ 7.43 (d, J=7.6 Hz, 1H), 7.29-7.26 (m, 2H), 7.25-7.23 (m, 1H), 7.20 (d, J= 8.0 Hz, 1H), 7.14 (d, J = 8.4 Hz, 2H), 6.90-6.85 (m, 3H), 6.72 (s, 1H), 4.39 (s, 3H), 4.19 (s, 2H), 4.01-3.96 (m, 2H), 3.74-3.64 (m, 1H), 3.39-3.36 (m, 6H), 3.19-3.15 (m, 4H), 2.86 (s, 6H); ESIMS: m/z = 540.1
[(M+H)+].
[0263] Example 38: Synthesis of 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)- 1 ,3 -dioxan- -yl)oxy)methyl)phenyl)pyridine.
Figure imgf000080_0002
To a stirred solution of l-(((2s,5s)-5-((4-bromobenzyl)oxy)-2-(2-chlorophenyl)-l,3- dioxan-2-yl)methyl)-lH-imidazole (0.25 g, 0.4 mmol) in 1 ,4-Dioxane/water (9: 1, 10 mL) was added K2CO3 (0.16 g, 1.2 mmol), pyridine-4-boronicacid pinacol ester (0.14 g, 0.7 mmol) and the reaction mixture was degassed with nitrogen gas for 10 minutes. Pd(dppf)Ci2 (0.029 g, 0.04 mmol) was added and again degassed with nitrogen gas for 10 minutes. The reaction mixture was then refluxed for 12 hours. The reaction mixture was filtered and concentrated and the residue was purified by prep TLC to afford the title compound. XH NMR: (400 MHz, CDC13) δ 8.65 (d, J = 5.2 Hz, 1H), 7.60 (d, J = 8.4 Hz, 2H), 7.51-7.43 (m, 3H), 7.36 (d, J = 8.4 Hz, 2H), 7.31-7.26 (m, 3H), 7.23-7.19 (m, 1H), 6.90 (s, 1H), 6.74 (s, 1H), 4.54 (s, 2H), 4.20 (s, 2H), 4.08-4.04 (m, 2H), 3.76-3.69 (m, 1H), 3.49-3.45 (m, 2H); ESIMS: m/z = 462.1 [(M+H)+].
[0264] Example 39: The compounds in the Table 23 below represent the extraction of over 200 compounds which realized the in vitro and in vivo goals. The target goals are defined in Table 22. As seen in Table 22 below, the in vitro goals are defined by efficacy targets: CYP17, CYP11, and CYP21. The off-target enzymes (where potency should be low) are CYP 19 and CYP3A4. Other parameters are no liver effects also estimated by bile acid synthesis inhibition.
Table 22: In vitro goals for compounds.
Figure imgf000081_0001
Table 23 : Representative examples of compounds of the disclosure and their potencies in Cyp l7, Cypl I, and Cyp21 assays.
Figure imgf000082_0002
[0265] Example 40: Pharmacokinetic studies in the guinea pig were run using lmg/kg IV dosing (20%DMA, 40% TEG, 40% water) and 10 mg/kg oral dosing (2% Tween-80, 98% HPMC (1 % water)). The oral PK data is summarized in the tables below.
Figure imgf000082_0001
FORMULATIONS [0266] Some embodiments of the present invention also relates to compositions or formulations which comprise the Cortisol lowering agents according to embodiments described herein. In general, the compositions of embodiments described herein comprise an effective amount of one or more compounds of the disclosure and salts thereof according to embodiments described herein which are effective for providing Cortisol lowering; and one or more excipients.
[0267] In this document, the term "excipient" and "carrier" are used interchangeably and said terms are defined herein as, "ingredients which are used in the practice of formulating a safe and effective pharmaceutical composition."
[0268] The formulator will understand that excipients are used primarily to serve in delivering a safe, stable, and functional pharmaceutical, serving not only as part of the overall vehicle for delivery but also as a means to achieve effective absorption by the recipient of the active ingredient. An excipient may fill a role as simple and direct as being an inert filler, or an excipient as used herein may be part of a pH stabilizing system or coating to insure delivery of the ingredients safely to the stomach. The formulator can also take advantage of the fact the compounds of embodiments described herein have improved cellular potency, pharmacokinetic properties, as well as improved oral bioavailability.
[0269] The present teachings also provide pharmaceutical compositions that include at least one compound described herein and one or more pharmaceutically acceptable carriers, excipients, or diluents. Examples of such carriers are well known to those skilled in the art and can be prepared in accordance with acceptable pharmaceutical procedures, such as, for example, those described in Remington 's Pharmaceutical Sciences, 17th edition, ed. Alfonoso R. Gennaro, Mack Publishing Company, Easton, PA (1985), the entire disclosure of which is incorporated by reference herein for all purposes. As used herein, "pharmaceutically acceptable" refers to a substance that is acceptable for use in pharmaceutical applications from a toxicological perspective and does not adversely interact with the active ingredient. Accordingly, pharmaceutically acceptable carriers are those that are compatible with the other ingredients in the formulation and are biologically acceptable. Supplementary active ingredients can also be incorporated into the pharmaceutical compositions.
[0270] Compounds of the present teachings can be administered orally or parenterally, neat or in combination with conventional pharmaceutical carriers. Applicable solid carriers can include one or more substances which can also act as flavoring agents, lubricants, solubilizers, suspending agents, fillers, glidants, compression aids, binders or tablet-disintegrating agents, or encapsulating materials. The compounds can be formulated in conventional manner. Oral formulations containing a compound disclosed herein can comprise any conventionally used oral form, including tablets, capsules, buccal forms, troches, lozenges and oral liquids, suspensions or solutions. In powders, the carrier can be a finely divided solid, which is an admixture with a finely divided compound. In tablets, a compound disclosed herein can be mixed with a carrier having the necessary compression properties in suitable proportions and compacted in the shape and size desired. The powders and tablets can contain up to 99 % of the compound.
[0271] Capsules can contain mixtures of one or more compound(s) disclosed herein with inert filler(s) and/or diluent(s) such as pharmaceutically acceptable starches (e.g., corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses (e.g., crystalline and microcrystalline celluloses), flours, gelatins, gums, and the like.
[0272] Useful tablet formulations can be made by conventional compression, wet granulation or dry granulation methods and utilize pharmaceutically acceptable diluents, binding agents, lubricants, disintegrants, surface modifying agents (including surfactants), suspending or stabilizing agents, including, but not limited to, magnesium stearate, stearic acid, sodium lauryl sulfate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, carboxymethylcellulose calcium, polyvinylpyrrolidine, alginic acid, acacia gum, xanthan gum, sodium citrate, complex silicates, calcium carbonate, glycine, sucrose, sorbitol, dicalcium phosphate, calcium sulfate, lactose, kaolin, mannitol, sodium chloride, low melting waxes, and ion exchange resins. Surface modifying agents include nonionic and anionic surface modifying agents. Representative examples of surface modifying agents include, but are not limited to, poloxamer 188, benzalkonium chloride, calcium stearate, cetostearl alcohol, cetomacrogol emulsifying wax, sorbitan esters, colloidal silicon dioxide, phosphates, sodium dodecylsulfate, magnesium aluminum silicate, and triethanolamine. Oral formulations herein can utilize standard delay or time-release formulations to alter the absorption of the compound(s). The oral formulation can also consist of administering a compound disclosed herein in water or fruit juice, containing appropriate solubilizers or emulsifiers as needed. [0273] Liquid carriers can be used in preparing solutions, suspensions, emulsions, syrups, elixirs, and for inhaled delivery. A compound of the present teachings can be dissolved or suspended in a pharmaceutically acceptable liquid carrier such as water, an organic solvent, or a mixture of both, or a pharmaceutically acceptable oils or fats. The liquid carrier can contain other suitable pharmaceutical additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers, and osmo-regulators. Examples of liquid carriers for oral and parenteral administration include, but are not limited to, water (particularly containing additives as described herein, e.g., cellulose derivatives such as a sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g., glycols) and their derivatives, and oils (e.g., fractionated coconut oil and arachis oil). For parenteral administration, the carrier can be an oily ester such as ethyl oleate and isopropyl myristate. Sterile liquid carriers are used in sterile liquid form compositions for parenteral administration. The liquid carrier for pressurized compositions can be halogenated hydrocarbon or other pharmaceutically acceptable propellants.
[0274] Liquid pharmaceutical compositions, which are sterile solutions or suspensions, can be utilized by, for example, intramuscular, intraperitoneal or subcutaneous injection. Sterile solutions can also be administered intravenously. Compositions for oral administration can be in either liquid or solid form.
[0275] Preferably the pharmaceutical composition is in unit dosage form, for example, as tablets, capsules, powders, solutions, suspensions, emulsions, granules, or suppositories. In such form, the pharmaceutical composition can be sub-divided in unit dose(s) containing appropriate quantities of the compound. The unit dosage forms can be packaged compositions, for example, packeted powders, vials, ampoules, prefilled syringes or sachets containing liquids. Alternatively, the unit dosage form can be a capsule or tablet itself, or it can be the appropriate number of any such compositions in package form. Such unit dosage form can contain from about 1 mg/kg of compound to about 500 mg/kg of compound, and can be given in a single dose or in two or more doses. Such doses can be administered in any manner useful in directing the compound(s) to the recipient's bloodstream, including orally, via implants, parenterally (including intravenous, intraperitoneal and subcutaneous injections), rectally, vaginally, and transdermally. [0276] When administered for the treatment or inhibition of a particular disease state or disorder, it is understood that an effective dosage can vary depending upon the particular compound utilized, the mode of administration, and severity of the condition being treated, as well as the various physical factors related to the individual being treated. In therapeutic applications, a compound of the present teachings can be provided to a patient already suffering from a disease in an amount sufficient to cure or at least partially ameliorate the symptoms of the disease and its complications. The dosage to be used in the treatment of a specific individual typically must be subjectively determined by the attending physician. The variables involved include the specific condition and its state as well as the size, age and response pattern of the patient.
[0277] In some cases it may be desirable to administer a compound directly to the airways of the patient, using devices such as, but not limited to, metered dose inhalers, breath-operated inhalers, multidose dry-powder inhalers, pumps, squeeze-actuated nebulized spray dispensers, aerosol dispensers, and aerosol nebulizers. For administration by intranasal or intrabronchial inhalation, the compounds of the present teachings can be formulated into a liquid composition, a solid composition, or an aerosol composition. The liquid composition can include, by way of illustration, one or more compounds of the present teachings dissolved, partially dissolved, or suspended in one or more pharmaceutically acceptable solvents and can be administered by, for example, a pump or a squeeze-actuated nebulized spray dispenser. The solvents can be, for example, isotonic saline or bacteriostatic water. The solid composition can be, by way of illustration, a powder preparation including one or more compounds of the present teachings intermixed with lactose or other inert powders that are acceptable for intrabronchial use, and can be administered by, for example, an aerosol dispenser or a device that breaks or punctures a capsule encasing the solid composition and delivers the solid composition for inhalation. The aerosol composition can include, by way of illustration, one or more compounds of the present teachings, propellants, surfactants, and co-solvents, and can be administered by, for example, a metered device. The propellants can be a chlorofluorocarbon (CFC), a hydrofluoroalkane (HFA), or other propellants that are physiologically and environmentally acceptable.
[0278] Compounds described herein can be administered parenterally or intraperitoneally. Solutions or suspensions of these compounds or a pharmaceutically acceptable salts, hydrates, or esters thereof can be prepared in water suitably mixed with a surfactant such as hydroxyl-propylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations typically contain a preservative to inhibit the growth of microorganisms.
[0279] The pharmaceutical forms suitable for injection can include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In some embodiments, the form can sterile and its viscosity permits it to flow through a syringe. The form preferably is stable under the conditions of manufacture and storage and can be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
[0280] Compounds described herein can be administered transdermally, i.e., administered across the surface of the body and the inner linings of bodily passages including epithelial and mucosal tissues. Such administration can be carried out using the compounds of the present teachings including pharmaceutically acceptable salts, hydrates, or esters thereof, in lotions, creams, foams, patches, suspensions, solutions, and suppositories (rectal and vaginal).
[0281] Transdermal administration can be accomplished through the use of a transdermal patch containing a compound, such as a compound disclosed herein, and a carrier that can be inert to the compound, can be non-toxic to the skin, and can allow delivery of the compound for systemic absorption into the blood stream via the skin. The carrier can take any number of forms such as creams and ointments, pastes, gels, and occlusive devices. The creams and ointments can be viscous liquid or semisolid emulsions of either the oil-in-water or water-in-oil type. Pastes comprised of absorptive powders dispersed in petroleum or hydrophilic petroleum containing the compound can also be suitable. A variety of occlusive devices can be used to release the compound into the blood stream, such as a semi-permeable membrane covering a reservoir containing the compound with or without a carrier, or a matrix containing the compound. Other occlusive devices are known in the literature.
[0282] Compounds described herein can be administered rectally or vaginally in the form of a conventional suppository. Suppository formulations can be made from traditional materials, including cocoa butter, with or without the addition of waxes to alter the suppository's melting point, and glycerin. Water-soluble suppository bases, such as polyethylene glycols of various molecular weights, can also be used.
[0283] Lipid formulations or nanocapsules can be used to introduce compounds of the present teachings into host cells either in vitro or in vivo. Lipid formulations and nanocapsules can be prepared by methods known in the art.
[0284] The compounds of embodiments described herein can be administered in the conventional manner by any route where they are active. Administration can be systemic, topical, or oral. For example, administration can be, but is not limited to, parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, oral, buccal, or ocular routes, or intravaginally, by inhalation, by depot injections, or by implants. Thus, modes of administration for the compounds of embodiments described herein (either alone or in combination with other pharmaceuticals) can be, but are not limited to, sublingual, injectable (including short-acting, depot, implant and pellet forms injected subcutaneous ly or intramuscularly), or by use of vaginal creams, suppositories, pessaries, vaginal rings, rectal suppositories, intrauterine devices, and transdermal forms such as patches and creams.
[0285] Specific modes of administration will depend on the indication. The selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response. The amount of compound to be administered is that amount which is therapeutically effective. The dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular animal treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician).
[0286] Pharmaceutical formulations containing the compounds of embodiments described herein and a suitable carrier can be solid dosage forms which include, but are not limited to, tablets, capsules, cachets, pellets, pills, powders and granules; topical dosage forms which include, but are not limited to, solutions, powders, fluid emulsions, fluid suspensions, semi-solids, ointments, pastes, creams, gels and jellies, and foams; and parenteral dosage forms which include, but are not limited to, solutions, suspensions, emulsions, and dry powder; comprising an effective amount of a polymer or copolymer of embodiments described herein. It is also known in the art that the active ingredients can be contained in such formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like. The means and methods for administration are known in the art and an artisan can refer to various pharmacologic references for guidance. For example, Modern Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Gilman's The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980) can be consulted.
[0287] The compounds of embodiments described herein can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. The compounds can be administered by continuous infusion subcutaneously over a period of about 15 minutes to about 24 hours. Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
[0288] For oral administration, the compounds can be formulated readily by combining these compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinylpyrrolidone (PVP). If desired, disintegrating agents can be added, such as, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
[0289] Dragee cores can be provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0290] Pharmaceutical preparations which can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
[0291] For buccal administration, the compositions can take the form of, e.g., tablets or lozenges formulated in a conventional manner.
[0292] For administration by inhalation, the compounds for use according to embodiments described herein are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
[0293] The compounds of embodiments described herein can also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
[0294] In addition to the formulations described previously, the compounds of embodiments described herein can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
[0295] Depot injections can be administered at about 1 to about 6 months or longer intervals. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. [0296] In transdermal administration, the compounds of embodiments described herein, for example, can be applied to a plaster, or can be applied by transdermal, therapeutic systems that are consequently supplied to the organism.
[0297] Pharmaceutical compositions of the compounds also can comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as, e.g., polyethylene glycols.
[0298] The compounds of embodiments described herein can also be administered in combination with other active ingredients, such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
[0299] In some embodiments, the disintegrant component comprises one or more of croscarmellose sodium, carmellose calcium, crospovidone, alginic acid, sodium alginate, potassium alginate, calcium alginate, an ion exchange resin, an effervescent system based on food acids and an alkaline carbonate component, clay, talc, starch, pregelatinized starch, sodium starch glycolate, cellulose floe, carboxymethylcellulose, hydroxypropylcellulose, calcium silicate, a metal carbonate, sodium bicarbonate, calcium citrate, or calcium phosphate.
[0300] In some embodiments, the diluent component comprises one or more of mannitol, lactose, sucrose, maltodextrin, sorbitol, xylitol, powdered cellulose, microcrystalline cellulose, carboxymethylcellulose, carboxyethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, methylhydroxyethylcellulose, starch, sodium starch glycolate, pregelatinized starch, a calcium phosphate, a metal carbonate, a metal oxide, or a metal aluminosilicate.
[0301] In some embodiments, the optional lubricant component, when present, comprises one or more of stearic acid, metallic stearate, sodium stearyl fumarate, fatty acid, fatty alcohol, fatty acid ester, glyceryl behenate, mineral oil, vegetable oil, paraffin, leucine, silica, silicic acid, talc, propylene glycol fatty acid ester, polyethoxylated castor oil, polyethylene glycol, polypropylene glycol, polyalkylene glycol, polyoxyethylene- glycerol fatty ester, polyoxyethylene fatty alcohol ether, polyethoxylated sterol, polyethoxylated castor oil, polyethoxylated vegetable oil, or sodium chloride. [0302] To increase the effectiveness of compounds of the present teachings, it can be desirable to combine a compound with other agents effective in the treatment of the target disease. For example, other active compounds (i.e., other active ingredients or agents) effective in treating the target disease can be administered with compounds of the present teachings. The other agents can be administered at the same time or at different times than the compounds disclosed herein.
[0303] Compounds of the present teachings can be useful for the treatment or inhibition of a pathological condition or disorder in a mammal, for example, a human subject. The present teachings accordingly provide methods of treating or inhibiting a pathological condition or disorder by providing to a mammal a compound of the present teachings inclding its pharmaceutically acceptable salt) or a pharmaceutical composition that includes one or more compounds of the present teachings in combination or association with pharmaceutically acceptable carriers. Compounds of the present teachings can be administered alone or in combination with other therapeutically effective compounds or therapies for the treatment or inhibition of the pathological condition or disorder.
[0304] Non-limiting examples of compositions according to embodiments described herein include from about 0.001 mg to about 1000 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 0.01 mg to about 100 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 100 mg to about 250 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 250 mg to about 500 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 500 mg to about 750 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; from about 750 mg to about 1000 mg of one or more compounds of the disclosure according to embodiments described herein and one or more excipients; and from about 0.1 mg to about 10 mg of one or more compounds of the disclosure according to embodiments described herein; and one or more excipients.
[0305] In some embodiments, the compositions according to embodiments described herein are administered orally to a patient once daily.
[0306] In some embodiments, the compositions according to embodiments described herein are administered orally to a patient twice daily. [0307] In some embodiments, the compositions according to embodiments described herein are administered orally to a patient three time per day.
[0308] In some embodiments, the compositions according to embodiments described herein are administered orally to a patient once weekly.
PROCEDURES
[0309] The following procedures can be utilized in evaluating and selecting compounds as Cortisol lowering agents.
[0310] Cypl7 assay protocol: AD293 cells that stably over-express recombinant CYP-17 were seeded in 96 well plates coated with poly D-lysine (15,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 μΐ, Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added. Compounds of the disclosure are then added to the wells in eight concentration spanning 10μΜ to 4.5 nM, and the plates are incubated for an additional 60 minutes at 37 °C. [21-3H] 17a-hydroxyl-Pregnenolone is then added (50 nCi per well, 31.25 nM) and the plates are incubated for an additional 4 hours at 37 °C. The media is then collected, 200 μΕ of chloroform is added, and the mixture is shaken for 1 hour. The aqueous layer is then separated and analyzed for the presence of 3H-acetic acid using a Perkin Elmer Topcount NXT to determine IC50S of the compounds of the disclosure.
[0311] Cyp21 assay protocol: AD293 cells that stably over-express recombinant CYP-21 were seeded in 96 well plates coated with poly D-lysine (10,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 μΐ, Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added. Compounds of the disclosure are then added to the wells in eight concentration spanning 10μΜ to 4.5 nM, and the plates are incubated for an additional 60 minutes at 37 °C. 17cc-OH Progesterone is then added (1.0 μΜ) and the plates are incubated for an additional 45 minutes at 37 °C. After incubation, 50 uL of the supernatant (medium) is transferred into a fresh plate and 150 uL of an acetonitrile solution containing 200ng/ml of Telmisartan is added. The sample is mixed and then placed in a centrifuge at 2000 rpm for 5 minutes. 100 uL of the supernatant is transferred into a fresh 96 well deep well plate, 100 uL of 1 : 1 methanol: water was added, the solution was mixed and then analyzed by LC/MS for the presence of 11-deoxycortisol using an Agilent 1200 RRLC/ABSCIEX API4000 LC-MS or Shimadzu Prominance/ABSCIEX API4000 LC-MS to determine IC50s of the compounds of the disclosure.
[0312] Cypll assay protocol: AD293 cells that stably over-express recombinant CYP- 11 were seeded in 96 well plates coated with poly D-lysine (15,000 cell per well) and incubated at 37 °C for 24 hours in Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment. The media is then removed, the cells are washed once with Phosphate buffer saline solution, and 50 Dulbecco's Modified Eagle Medium (DMEM) with Fetal Bovine Serum (FBS) that is stripped of hormones by charcoal treatment is added. Compounds of the disclosure are then added to the wells in eight concentration spanning 10μΜ to 4.5 nM, and the plates are incubated for an additional 60 minutes at 37 °C. l 1-deoxycortisol is then added (2.0 μΜ) and the plates are incubated for an additional 12 hours at 37 °C. After incubation, 50 uL of the supernatant (medium) is transferred into a fresh plate and 150 uL of an acetonitrile solution containing 200ng/ml of Telmisartan is added. The sample is mixed and then placed in a centrifuge at 2000 rpm for 5 minutes. 100 uL of the supernatant is transferred into a fresh 96 well deep well plate, 100 uL of 1 : 1 methanol: water was added, the solution was mixed and then analyzed by LC/MS for the presence of Cortisol using an Agilent 1200 RRLC/ABSCIEX API4000 LC-MS or Shimadzu Prominance/ABSCIEX API4000 LC-MS to determine IC50S of the compounds of the disclosure.
[0313] Results for representative compounds according to embodiments described herein are listed in Table 23.
Table 23: Representative examples of compounds of the disclosure and their potencies in Cypl7, Cypl 1, and Cyp21 assays.
Figure imgf000094_0002
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001

Claims

WHAT IS CLAIMED IS:
1. A compound comprisin formula (I):
Figure imgf000099_0001
including hydrates, solvates, enantiomers, diastereomers, pharmaceutically acceptable salts, prodrugs and complexes thereof, wherein:
Q is selected from a group consisting of optionally substituted aryl, optionally substituted heteroaryl, \— / , and \— / ;
Rla, Rlb, Rlc, Rld, and Rle are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted C e linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6, optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7and -S02NHR6; R2a, R2b, R2c, R2d, R2e, R2f and R2g are each independently selected from the group consisting of hydrogen, halogen, OH, optionally substituted Ci_6 linear alkyl, optionally substituted C e branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, Ci_6 optionally substituted alkoxy, -NR4aR4b, -NR5COR6, -C02R6, -C02NR4aR4b, -NHS02R7, -SH, -SR7, S02R7 and -S02NHR6;
3 is selected from a group consisting of -S02R8, -C(0)NR9R10, -C(0)OR7,
Figure imgf000099_0002
R a and R are each independently selected from the group consisting of hydrogen, optionally substituted C e linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl;
R5 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl; R6 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, and optionally substituted C3-7 cycloalkyl;
R7 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci-6 branched alkyl, and optionally substituted C3-7 cycloalkyl;
R8 is selected from the group consisting of optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7 cycloalkyl, optionally substituted Ci_6 haloalkyl, optionally substituted aryl, optionally substituted heteroaryl, and optionally substituted C3-7 heterocyclyl;
R9 is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted C3-7
cycloalkyl, optionally substituted Ci_6 haloalky
Figure imgf000100_0001
R is selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, and optionally substituted Ci_6 branched alkyl;
Rlla and Rl lb are each independently selected from the group consisting of hydrogen, optionally substituted Ci_6 linear alkyl, optionally substituted Ci_6 branched alkyl, optionally substituted aryl, optionally substituted benzyl, - CH2OR6, and CH2Heteroaryl.
2. The compound of claim 1, wherein the compound comprises formula (II):
Figure imgf000100_0002
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
The compound of claim 1, having the formula (III):
Figure imgf000101_0001
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
4. The compound of claim 1, having the formula (IV):
Figure imgf000101_0002
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
5. The com ound of claim 1, having formula (V):
Figure imgf000101_0003
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
6. The compound of claim 1, having formula (VI):
Figure imgf000102_0001
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
7. The compound of claim 1, having formula (VII):
Figure imgf000102_0002
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
The compound of claim 1, having formula (VIII):
Figure imgf000102_0003
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
9. The compound of claim 1, having formula (IX):
Figure imgf000102_0004
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
10. The compound of claim 1, having formula (X):
Figure imgf000103_0001
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
11. The compound of claim 1 , having formula (Xa):
Figure imgf000103_0002
a hydrate, a solvate, an enantiomer, a diastereomer, a pharmaceutically acceptable salt, a prodrug or a complex thereof.
12. A compound of the claim 1 that is:
1 -(4-(4-((((2s,5s)-2-(( lH-imidazol- 1 -yl)methyl)-2-(2,4-dichlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine:
3-((4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)sulfonyl)benzonitrile;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-chloropropyl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(cyclopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(isopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(ethylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((lH-imidazol-4-yl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-
(trifluoromethoxy)phenyl)sulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(pyridin-3-ylsulfonyl)piperazine;
l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
1 -(4-(4-((((2r,5r)-2-((l H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)pyridine;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)morpholine;
4- (4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)morpholine;
1- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
1 -(4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
2- methoxyethyl 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; 2-methoxyethyl 4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate; ethyl 2-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine- 1 -carboxamide;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)pyridine;
or a pharmaceutically acceptable salt form thereof.
13. A composition comprising an effective amount of at least one compound
according to claim 1.
14. A composition according to claim 13, further comprising at least one
excipient.
15. A composition according to claim 14, wherein the at least one compound is selected from the group consisting of:
1 -(4-(4-((((2s,5s)-2-(( lH-imidazol- 1 -yl)methyl)-2-(2,4-dichlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine:
3-((4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)sulfonyl)benzonitrile;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-chloropropyl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(cyclopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(isopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(ethylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((lH-imidazol-4-yl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-
(trifluoromethoxy)phenyl)sulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(pyridin-3-ylsulfonyl)piperazine;
l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
1 -(4-(4-((((2r,5r)-2-((l H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)pyridine;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)morpholine;
4- (4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)morpholine;
1- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
1 -(4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
2- methoxyethyl 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; 2-methoxyethyl 4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate;
ethyl 2-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine- 1 -carboxamide;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)pyridine;
a pharmaceutically acceptable salt form thereof, or a combination thereof.
16. A method of treating a disease associated with overproduction of Cortisol, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease.
17. The method of claim 16, wherein the at least one compound is administered in a composition further comprising at least one excipient.
18. The method of claim 17, wherein the at least one compound is at least one member selected from the group consisting of:
1 -(4-(4-((((2s,5s)-2-(( lH-imidazol- 1 -yl)methyl)-2-(2,4-dichlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine:
3-((4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazin-l-yl)sulfonyl)benzonitrile;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-chloropropyl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(cyclopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(isopropylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(ethylsulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((lH-imidazol-4-yl)sulfonyl)piperazine; l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-((3-
(trifluoromethoxy)phenyl)sulfonyl)piperazine;
l-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)-4-(pyridin-3-ylsulfonyl)piperazine;
l-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone; 1 -(4-(4-((((2r,5r)-2-((l H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 - dioxan-5 -yl)oxy)methyl)phenyl)piperazin- 1 -yl)ethanone;
4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2,4-dichlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)pyridine;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)morpholine;
4- (4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)morpholine;
1- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
1 -(4-((((2r,5r)-2-(( 1 H-imidazol- 1 -yl)methyl)-2-(2-chlorophenyl)- 1 ,3 -dioxan-5 - yl)oxy)methyl)phenyl)-4-(methylsulfonyl)piperazine;
2- methoxyethyl 4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; 2-methoxyethyl 4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2- chlorophenyl)-l,3-dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxylate; ethyl 2-(4-(4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate;
ethyl 2-(4-(4-((((2r,5r)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3- dioxan-5-yl)oxy)methyl)phenyl)piperazine-l-carboxamido)acetate;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)-N,N-dimethylpiperazine- 1 -carboxamide;
4- (4-((((2s,5s)-2-((lH-imidazol-l-yl)methyl)-2-(2-chlorophenyl)-l,3-dioxan-
5- yl)oxy)methyl)phenyl)pyridine;
a pharmaceutically acceptable salt form thereof, or a combination thereof.
19. The method of claim 16, wherein the disease associated with overproduction of Cortisol is metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke or incidentalomas.
20. The method of claim 17, wherein the disease associated with overproduction of Cortisol is metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke or incidentalomas.
21. A method of treating a disease associated with excess Cypl7 activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease.
22. The method of claim 21, wherein the at least one compound is administered in a composition further comprising at least one excipient.
23. A method of treating a disease associated with excess Cypl lBl activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease.
24. The method of claim 23, wherein the at least one compound is administered in a composition further comprising at least one excipient.
25. A method of treating a disease associated with excess Cyp21 activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease.
26. The method of claim 25, wherein the at least one compound is administered in a composition further comprising at least one excipient.
27. A method of treating a disease associated with Cypl7 activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease, wherein the Cypl7 activity is lowered.
28. The method of claim 27, wherein the at least one compound is administered in a composition further comprising at least one excipient.
29. A method of treating a disease associated with Cypl 1B1 activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease wherein Cypl IB 1 activity is lowered.
30. The method of claim 29, wherein the at least one compound is administered in a composition further comprising at least one excipient.
31. A method of treating a disease associated with Cyp21 activity, said method comprising administering to a subject an effective amount of at least one compound according to claim 1 to treat the disease wherein Cyp21 activity is lowered.
32. The method of claim 31, wherein the at least one compound is administered in a composition further comprising at least one excipient.
33. A method of treating a disease selected from metabolic syndrome, obesity, headache, depression, hypertension, diabetes mellitus, Cushing's Syndrome, pseudo-Cushing syndrome, cognitive impairment, dementia, heart failure, renal failure, psoriasis, glaucoma, cardiovascular disease, stroke, or incidentalomas, the method comprising administering to a subject an effective amount of at least one compound according to the claim 1 to treat the disease, wherein the at least one compound lowers the activity of at least two of the following: Cypl7, Cyp21, or Cypl lBl .
34. The method of claim 33, wherein the at least one compound modulates Cortisol activity.
PCT/US2014/057490 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition WO2015048311A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA2925294A CA2925294A1 (en) 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition
US15/024,766 US20160244436A1 (en) 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibiting cytochrome p450 inhibition and their method of use
EP14847772.2A EP3049084A4 (en) 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition
CN201480063953.0A CN105764512A (en) 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibiting cytochrome P450 inhibition
JP2016516909A JP2016536273A (en) 2013-09-25 2014-09-25 Novel functionalized 5- (phenoxymethyl) -1,3-dioxane analogs exhibiting cytochrome P450 inhibition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361882625P 2013-09-25 2013-09-25
US61/882,625 2013-09-25

Publications (1)

Publication Number Publication Date
WO2015048311A1 true WO2015048311A1 (en) 2015-04-02

Family

ID=52744463

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/057490 WO2015048311A1 (en) 2013-09-25 2014-09-25 Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition

Country Status (6)

Country Link
US (1) US20160244436A1 (en)
EP (1) EP3049084A4 (en)
JP (1) JP2016536273A (en)
CN (1) CN105764512A (en)
CA (1) CA2925294A1 (en)
WO (1) WO2015048311A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017070719A2 (en) 2015-10-22 2017-04-27 Mangosuthu University Of Technology Pharmacophores, compounds and methods having application in the treatment of cancer through inhibition of cyp17a1 and cyp19a1

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4144346A (en) * 1977-01-31 1979-03-13 Janssen Pharmaceutica N.V. Novel 1-(1,3-dioxolan-2-ylmethyl)-1H-imidazoles
US4503055A (en) * 1979-03-26 1985-03-05 Janssen Pharmaceutica, N.V. Derivatives of [4-(piperazin-1-yl-phenyloxymethyl)-1,3-dioxolan-2-ylmethyl]-1H-imidazoles and 1H-1,2,4-triazoles
US20050013834A1 (en) * 2001-07-26 2005-01-20 Ljusberg Barbro Helena Pharmaceutical formulations comprising ketoconazole
WO2008089461A1 (en) * 2007-01-18 2008-07-24 Evolva Sa Substituted 1,3-dioxanes useful as ppar modulators
US20090238473A1 (en) * 2008-03-19 2009-09-24 Honeywell International Inc. Construction of evidence grid from multiple sensor measurements
US20100048562A1 (en) * 2006-12-18 2010-02-25 Christopher Adams Organic compounds
US20100093755A1 (en) * 2006-10-02 2010-04-15 Cortendo Invest Ab, Ketoconazole Enantiomer in Humans
US20110105522A1 (en) * 2008-02-29 2011-05-05 Sridhar Mani Ketoconazole-derivative antagonist of human pregnane x receptor and uses thereof
US20110112067A1 (en) * 2009-11-09 2011-05-12 Universitat Des Saarlandes Inhibitors of the Human Aldosterone Sythase CYP11B2
US20110160154A1 (en) * 2007-06-13 2011-06-30 Auspex Pharmaceuticals, Inc. Substituted piperazines
WO2012052540A1 (en) * 2010-10-21 2012-04-26 Universitaet Des Saarlandes Selective cyp11b1 inhibitors for the treatment of cortisol dependent diseases
WO2012083112A2 (en) * 2010-12-16 2012-06-21 Biomarin Pharmaceutical Inc. Cyp11b, cyp17, and/or cyp21 inhibitors

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4144346A (en) * 1977-01-31 1979-03-13 Janssen Pharmaceutica N.V. Novel 1-(1,3-dioxolan-2-ylmethyl)-1H-imidazoles
US4503055A (en) * 1979-03-26 1985-03-05 Janssen Pharmaceutica, N.V. Derivatives of [4-(piperazin-1-yl-phenyloxymethyl)-1,3-dioxolan-2-ylmethyl]-1H-imidazoles and 1H-1,2,4-triazoles
US20050013834A1 (en) * 2001-07-26 2005-01-20 Ljusberg Barbro Helena Pharmaceutical formulations comprising ketoconazole
US20100093755A1 (en) * 2006-10-02 2010-04-15 Cortendo Invest Ab, Ketoconazole Enantiomer in Humans
US20100048562A1 (en) * 2006-12-18 2010-02-25 Christopher Adams Organic compounds
WO2008089461A1 (en) * 2007-01-18 2008-07-24 Evolva Sa Substituted 1,3-dioxanes useful as ppar modulators
US20110160154A1 (en) * 2007-06-13 2011-06-30 Auspex Pharmaceuticals, Inc. Substituted piperazines
US20110105522A1 (en) * 2008-02-29 2011-05-05 Sridhar Mani Ketoconazole-derivative antagonist of human pregnane x receptor and uses thereof
US20090238473A1 (en) * 2008-03-19 2009-09-24 Honeywell International Inc. Construction of evidence grid from multiple sensor measurements
US20110112067A1 (en) * 2009-11-09 2011-05-12 Universitat Des Saarlandes Inhibitors of the Human Aldosterone Sythase CYP11B2
WO2012052540A1 (en) * 2010-10-21 2012-04-26 Universitaet Des Saarlandes Selective cyp11b1 inhibitors for the treatment of cortisol dependent diseases
WO2012083112A2 (en) * 2010-12-16 2012-06-21 Biomarin Pharmaceutical Inc. Cyp11b, cyp17, and/or cyp21 inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHAPMAN, DR ET AL.: "Synthesis of Diastereomeric Ketoconazole Analogs", J. HETEROCYCLIC CHEM., vol. 27, 1990, pages 2063 - 2068, XP055125995 *
HU, Q ET AL.: "Replacement of Imidazolyl by Pyridyl in Biphenylmethylenes Results in Selective CYP17 and Dual CYP17/CYP11B1 Inhibitors for the Treatment of Prostate Cancer", JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, 15 July 2010 (2010-07-15), pages 5749 - 5758, XP055334507 *
See also references of EP3049084A4 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017070719A2 (en) 2015-10-22 2017-04-27 Mangosuthu University Of Technology Pharmacophores, compounds and methods having application in the treatment of cancer through inhibition of cyp17a1 and cyp19a1
CN108701171A (en) * 2015-10-22 2018-10-23 马古苏托科技大学 In pharmacophore, the Compounds and methods for by inhibiting that there is application in CYP17A1 and CYP19A1 treating cancers
CN108779085A (en) * 2015-10-22 2018-11-09 马古苏托科技大学 In pharmacophore, the Compounds and methods for by inhibiting that there is application in CYP17A1 and CYP19A1 treating cancers
EP3365330A4 (en) * 2015-10-22 2019-06-05 Mangosuthu University Of Technology Pharmacophores, compounds and methods having application in the treatment of cancer through inhibition of cyp17a1 and cyp19a1
EP3365819A4 (en) * 2015-10-22 2019-07-24 Mangosuthu University Of Technology Pharmacophores, compounds and methods having application in the treatment of cancer through inhibition of cyp17a1 and cyp19a1
CN108779085B (en) * 2015-10-22 2022-03-22 马古苏托科技大学 Pharmacophores, compounds and methods having application in the treatment of cancer by inhibition of CYP17a1 and CYP19a1
CN108701171B (en) * 2015-10-22 2022-06-10 马古苏托科技大学 Pharmacophores, compounds and methods having application in the treatment of cancer by inhibition of CYP17a1 and CYP19a1

Also Published As

Publication number Publication date
EP3049084A4 (en) 2017-03-15
CA2925294A1 (en) 2015-04-02
EP3049084A1 (en) 2016-08-03
JP2016536273A (en) 2016-11-24
US20160244436A1 (en) 2016-08-25
CN105764512A (en) 2016-07-13

Similar Documents

Publication Publication Date Title
AU2013226013B2 (en) Inhibitors of Hepatitis B Virus Covalently Closed Circular DNA Formation and Their Method of Use
JP4493503B2 (en) Heterocyclic compounds and antitumor agents containing the same as active ingredients
US11897870B2 (en) 5-hydroxytryptamine receptor 7 activity modulators and their method of use
US9453014B2 (en) Cyclic amide derivatives as inhibitors of 11-β-hydroxysteroid dehydrogenase and uses thereof
MX2007008757A (en) Substituted triazole derivatives as oxytocin antagonists.
US20150337003A1 (en) Abiraterone and analogs thereof for the treatment of diseases associated with cortisol overproduction
US20220133713A1 (en) Sigma-2 receptor binders and their method of use
EP3200589A1 (en) Novel 5-hydroxytryptamine receptor 7 activity modulators and their method of use
US9303016B2 (en) Derivatives of aza adamantane and uses thereof
BR112013014966B1 (en) CONTINUOUS ARICYCLIC COMPOUND, DIACYLGLYCEROL ACYLTRANSPHERASE INHIBITOR (DGAT) 1 AND ITS USES
US9725436B2 (en) Cytochrome P450 inhibitors and their method of use
WO2015048311A1 (en) Novel functionalized 5-(phenoxymethyl)-1,3-dioxane analogs exhibitng cytochrome p450 inhibition
WO2014180544A1 (en) Hydantoine derivatives as cd38 inhibitors
WO2015112369A1 (en) Novel cytochrome p450 inhibitors and their method of use
WO2014122530A1 (en) Novel functionalized 4-(phenoxymethyl(-1,3-dioxolane analogs exhibiting cytochrome p450 inhibition and their method of use
WO2015054117A1 (en) Functionalized furan-2-sulfonamides exhibiting endothelial lipase inhibition
WO2000078758A1 (en) Novel imidazole derivatives

Legal Events

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

Ref document number: 14847772

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2925294

Country of ref document: CA

Ref document number: 2016516909

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2014847772

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014847772

Country of ref document: EP