WO2008124665A1 - Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof - Google Patents

Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof Download PDF

Info

Publication number
WO2008124665A1
WO2008124665A1 PCT/US2008/059528 US2008059528W WO2008124665A1 WO 2008124665 A1 WO2008124665 A1 WO 2008124665A1 US 2008059528 W US2008059528 W US 2008059528W WO 2008124665 A1 WO2008124665 A1 WO 2008124665A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
benzenesulfonamide
ethyl
trifluoro
trimethyl
Prior art date
Application number
PCT/US2008/059528
Other languages
French (fr)
Inventor
Hossein Razavi
Daniel Kuzmich
Original Assignee
Boehringer Ingelheim International Gmbh
Boehringer Ingelheim Pharma Gmbh & Co. Kg
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 Boehringer Ingelheim International Gmbh, Boehringer Ingelheim Pharma Gmbh & Co. Kg filed Critical Boehringer Ingelheim International Gmbh
Priority to JP2010503136A priority Critical patent/JP2010523689A/en
Priority to CA002683647A priority patent/CA2683647A1/en
Priority to US12/520,997 priority patent/US20100063282A1/en
Priority to EP08745203A priority patent/EP2136634A4/en
Publication of WO2008124665A1 publication Critical patent/WO2008124665A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • C07D209/48Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/84Nitriles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/233Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/24Oxygen atoms attached in position 8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • C07D215/40Nitrogen atoms attached in position 8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D219/00Heterocyclic compounds containing acridine or hydrogenated acridine ring systems
    • C07D219/04Heterocyclic compounds containing acridine or hydrogenated acridine ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • C07D219/06Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/04Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D233/28Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/44Nitrogen atoms not forming part of a nitro radical
    • C07D233/46Nitrogen atoms not forming part of a nitro radical with only hydrogen atoms attached to said nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/38One sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/64Benzothiazoles with only hydrocarbon or substituted hydrocarbon radicals attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/82Benzo [b] furans; Hydrogenated benzo [b] furans with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • C07D307/83Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/14Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted in position 6 and unsubstituted in position 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/101,4-Dioxanes; Hydrogenated 1,4-dioxanes
    • C07D319/141,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems
    • C07D319/161,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D319/18Ethylenedioxybenzenes, not substituted on the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D321/00Heterocyclic compounds containing rings having two oxygen atoms as the only ring hetero atoms, not provided for by groups C07D317/00 - C07D319/00
    • C07D321/02Seven-membered rings
    • C07D321/10Seven-membered rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/54Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • 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
    • C07D405/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to glucocorticoid mimetics or ligands, methods of making such compounds, their use in pharmaceutical compositions, and their use in modulating the glucocorticoid receptor function, treating disease-states or conditions mediated by the glucocorticoid receptor function in a patient in need of such treatment, and other uses.
  • Glucocorticoids a class of corticosteroids, are endogenous hormones with profound effects on the immune system and multiple organ systems. They suppress a variety of immune and inflammatory functions by inhibition of inflammatory cytokines such as IL-I, IL-2, IL-6, and TNF, inhibition of arachidonic acid metabolites including prostaglandins and leukotrienes, depletion of T-lymphocytes, and reduction of the expression of adhesion molecules on endothelial cells (PJ. Barnes, Clin. ScL, 1998, 94, pp. 557-572; P.J. Barnes et al, Trends Pharmacol. ScL, 1993, J_4, pp. 436-441). In addition to these effects, glucocorticoids stimulate glucose production in the liver and catabolism of proteins, play a role in electrolyte and water balance, reduce calcium absorption, and inhibit osteoblast function.
  • IL-I IL-I
  • IL-2 interleukin-2
  • IL-6 interle
  • glucocorticoids The anti-inflammatory and immune suppressive activities of endogenous glucocorticoids have stimulated the development of synthetic glucocorticoid derivatives including dexamethasone, prednisone, and prednisolone (L. Parente, Glucocorticoids. N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, pp. 35-54).
  • rheumatic diseases such as rheumatoid arthritis, juvenile arthritis, and ankylosing spondylitis
  • dermatological diseases including psoriasis and pemphigus
  • allergic disorders including allergic rhinitis, atopic dermatitis, and contact dermatitis
  • pulmonary conditions including asthma and chronic obstructive pulmonary disease (COPD)
  • COPD chronic obstructive pulmonary disease
  • Crohn disease ulcerative colitis
  • systemic lupus erythematosus autoimmune chronic active hepatitis
  • osteoarthritis tendonitis
  • bursitis J. Toogood, Glucocorticoids, N.J.
  • glucocorticoid receptor The effects of glucocorticoids are mediated at the cellular level by the glucocorticoid receptor (R.H. Oakley and J. Cidlowski, Glucocorticoids, N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, pp. 55-80).
  • the glucocorticoid receptor is a member of a class of structurally related intracellular receptors that when coupled with a ligand can function as a transcription factor that affects gene expression (R.M. Evans, Science, 1988, 240, pp. 889- 895).
  • Other members of the family of steroid receptors include the mineralocorticoid, progesterone, estrogen, and androgen receptors.
  • glucocorticoids N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, is hereby incorporated by reference in its entirety to better describe the state of the art.
  • transactivation the translocation of the ligand-bound glucocorticoid receptor to the nucleus is followed by binding to glucocorticoid response elements (GREs) in the promoter region of side effect- associated genes, for example, phosphoenolpyruvate carboxy kinase (PEPCK) in the case of increased glucose production.
  • GREs glucocorticoid response elements
  • PEPCK phosphoenolpyruvate carboxy kinase
  • the anti-inflammatory effects are thought to be due to a process called transrepression.
  • transrepression is a process independent of DNA binding that results from inhibition of NF -kB and AP-I- mediated pathways, leading to down regulation of many inflammatory and immune mediators. Additionally, it is believed that a number of the observed side effects may be due to the cross-reactivity of the currently available glucocorticoids with other steroid receptors, particularly the mineralocorticoid and progesterone receptors.
  • ligands for the glucocorticoid receptor that are highly selective and, upon binding, can dissociate the transactivation and transrepression pathways, providing therapeutic agents with a reduced side effect profile.
  • Assay systems to determine effects on transactivation and transrepression have been described (e.g., CM. Bamberger and H.M. Schulte, Eur. J. Clin. Invest., 2000, 30 (suppl. 3), pp. 6-9).
  • Selectivity for the glucocorticoid receptor may be determined by comparing the binding affinity for this receptor with that of other steroid family receptors including those mentioned above.
  • Glucocorticoids also stimulate the production of glucose in the liver by a process called gluconeogenesis and it is believed that this process is mediated by transactivation events. Increased glucose production can exacerbate type II diabetes, therefore a compound that selectivity inhibited glucocorticoid mediated glucose production may have therapeutic utility in this indication (J.E. Freidman et al, J. Biol. Chem., 1997, 272, pp. 31475-31481).
  • PCT International Publication No. WO 99/33786 discloses triphenylpropanamide compounds with potential use in treating inflammatory diseases.
  • PCT International Publication No. WO 00/66522 describes non-steroidal compounds as selective modulators of the glucocorticoid receptor potentially useful in treating metabolic and inflammatory diseases.
  • PCT International Publication No. WO 99/41256 describes tetracyclic modulators of the glucocorticoid receptor potentially useful in treating immune, autoimmune, and inflammatory diseases.
  • U.S. Patent No. 5,688,810 describes various non-steroidal compounds as modulators of glucocorticoid and other steroid receptors.
  • PCT International Publication No. WO 99/63976 describes a non-steroidal, liver-selective glucocorticoid antagonist potentially useful in the treatment of diabetes.
  • PCT International Publication No. WO 00/32584 discloses non-steroidal compounds having anti-inflammatory activity with dissociation between anti-inflammatory and metabolic effects.
  • PCT International Publication No. WO 98/54159 describes non-steroidal cyclically substituted acylanilides with mixed gestagen and androgen activity.
  • U.S. Patent No. 4,880,839 describes acylanilides having progestational activity and EP 253503 discloses acylanilides with antiandrogenic properties.
  • PCT International Publication No. WO 97/27852 describes amides that are inhibitors of farnesylprotein transferase.
  • a compound that is found to interact with the glucocorticoid receptor in a binding assay could be an agonist or an antagonist.
  • the agonist properties of the compound could be evaluated in the transactivation or transrepression assays described above.
  • the compound may be found to have antagonist activity.
  • glucocorticoids stimulate glucose production in the liver.
  • a ligand for the glucocorticoid receptor that is found to be an antagonist may be useful, inter alia, for treating or preventing diabetes.
  • the instant invention is directed to compounds of Formula (IA)
  • R 1 is hydrogen or Ci-C 3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo;
  • R 2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups, wherein each substituent group of R 2 is independently C 1 -Cs alkyl, C 2 -C3 alkenyl, C 2 - C3 alkynyl, C 1 -Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C 1 -Cs alkoxycarbonylamino, C 1 -Cs alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C 1 -
  • each substituent group of R 2 is optionally independently substituted with C 1 -C 3 alkyl, halogen, hydroxyl, or amino,
  • R 2 cannot be /?-methylphenyl
  • R 3 is a hydrogen or C 1 -C 5 alkyl, optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 3 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
  • R 4 and R 5 are each independently hydrogen, C 1 -C 5 alkyl, or phenyl, or R 4 and R 5 together with the carbon atom they are commonly attached to form a C 3 -C 8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 4 and R 5 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
  • R 6 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups
  • each substituent group of R 6 is independently C 1 -C 3 alkyl, C 2 -C 5 alkenyl, C 2 -
  • each substituent group of R 6 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C 3 alkyl, Ci-C 3 alkoxy, halogen, hydroxy, oxo, cyano, phenyl, amino, or trifluoromethyl;
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, Ci-C 5 alkyl, or phenyl
  • each substituent group of R 7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C 3 alkyl, Ci-C 3 alkoxy, phenyl optionally substituted with Ci-C 3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl,
  • One aspect of the invention includes compounds of Formula (IA), wherein:
  • R 1 is hydrogen
  • R 2 is phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
  • each substituent group of R 2 is independently Ci-C 3 alkyl, C 1 -Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C 1 -Cs alkyl, or Ci-C 5 alkylthio, wherein R 2 cannot be p-methylphenyl;
  • R 3 is hydrogen
  • R 4 and R 5 are each hydrogen or C 1 -Cs alkyl
  • R 6 is an indolyl, dihydroisoindolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, dihydroisobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, dihydrobenzodioxepinyl, acridinyl, pyrimidinyl or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 6 is independently Ci-C 3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C 1 -C 3 alkoxy, acylamino, aminocarbonyl, C 1 -C 3 alkylaminocarbonyl, C 1 -C 3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or Ci-C 3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
  • each substituent group of R 6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, phenyl, or trifluoromethyl;
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, Ci-C 5 alkyl, or phenyl
  • each substituent group of R 7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C 3 alkyl, Ci-C 3 alkoxy, phenyl, hydroxy, oxo, cyano, amino, or trifluoromethyl, or a tautomer, prodrug, solvate, or salt thereof.
  • Another aspect of the invention includes compounds of Formula (IA), wherein:
  • R 1 is hydrogen
  • R 2 is a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
  • each substituent group of R 2 is independently C 1 -Cs alkyl, C 1 -C 3 alkoxy, hydroxy, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C 1 -C 3 alkyl, or Ci-C 3 alkylthio,
  • R 2 cannot be />-methylphenyl
  • R 3 is hydrogen
  • R 4 and R 5 are each hydrogen
  • R 6 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 6 is independently Ci-C 3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, Ci-C 3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or Ci-C 3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, wherein each substituent group of R 6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl;
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, C 1 -C 5 alkyl, or phenyl
  • Preferred compounds of Formula (IA) include the following:
  • More preferred compounds of Formula (IA) include the following:
  • the invention also provides a method of making a compound of Formula (IA)
  • R 1 is H and R 2 , R 3 , R 4 , R 5 , R 6 , and X are as defined above, the method comprising reacting an aziridine compound of Formula (II) with a reagent R 6 X-M of Formula (III) where M is Na, K, or Li, or where X is nitrogen or sulfur and M is hydrogen, in a suitable solvent to form the compound of Formula (IA)
  • the invention further provides a method of making a compound of Formula (IA)
  • R 1 , R 3 , R 4 , and R 5 are each H, and R 2 and R 6 are as defined above, the method comprising:
  • a second method for making a compound of Formula (IA) comprises:
  • the group R 2 may be substituted with another R 2 group, the method comprising:
  • the invention is also directed to compounds of Formula (IB)
  • R 1 is hydrogen or Ci-C 3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo;
  • R 2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups
  • each substituent group of R 2 is independently Ci-C 5 alkyl, C 2 -C 3 alkenyl, C 2 - C 3 alkynyl, C 1 -C5 alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C 1 -C5 alkoxycarbonylamino, C 1 -C5 alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C 1 - C 5 alkyl; or ureido wherein either nitrogen atom is optionally independently substituted with C 1 -C5 alkyl; or C 1 -C5 alkylthio,
  • each substituent group of R is optionally independently substituted with Ci- C 3 alkyl, halogen, hydroxyl, or amino, wherein R 2 cannot be p-methylphenyl;
  • R 3 is Ci-Cs alkyl independently substituted with one, two, three, four, or five substituent groups,
  • each substituent group of R 3 is independently C 3 -C 6 cycloalkyl, aryl, halogen, trifluoromethyl, trifluoromethoxy, or trifluoromethylthio,
  • R cannot be a trifluoromethyl
  • R 4 is a hydrogen or C 1 -Cs alkyl, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 4 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
  • R 4 cannot be a trifluoromethyl
  • R 5 and R 6 are each independently hydrogen, C 1 -Cs alkyl or phenyl or R 5 and R 6 together with the carbon atom they are commonly attached to form a C3-C8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 5 and R 6 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, Ci-C 5 alkyl, or phenyl
  • each substituent group of R 7 is optionally independently substituted with one, two, or three substituent groups selected from C 1 -C 3 alkyl, C 1 -C 3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and R 8 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 8 is independently C 1 -C 3 alkyl, C 2 -C 5 alkenyl, C 2 -
  • each substituent group of R 8 is optionally independently substituted with one, two, or three substituent groups selected from C 1 -C 3 alkyl, C 1 -C 3 alkoxy, halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
  • One aspect of the invention includes compounds of Formula (IB), wherein:
  • R 1 is hydrogen
  • R 2 is phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
  • each substituent group of R 2 is independently C 1 -C 3 alkyl, C 1 -Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C 1 -Cs alkyl, or Ci-C 5 alkylthio,
  • R 2 cannot be /?-methylphenyl
  • R 3 is Ci-C 5 alkyl independently substituted with one, two, three, four, or five substituent groups,
  • each substituent group of R 3 is independently C 3 -C 8 cycloalkyl, halogen, trifluoromethyl, or trifluoromethoxy,
  • R 3 cannot be a trifluoromethyl
  • R 4 is hydrogen
  • R 5 and R 6 are each hydrogen or C 1 -Cs alkyl
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, C 1 -C 5 alkyl, or phenyl
  • each substituent group of R 7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C 3 alkyl, Ci-C 3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and
  • R 8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 8 is independently Ci-C 3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, Ci-C 3 alkoxy, acylamino, aminocarbonyl, Ci-C 3 alkylaminocarbonyl, Ci-C 3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, trifluoromethyl, or Ci-C 3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, wherein each substituent group of R 8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl ,
  • Another aspect of the invention includes compounds of Formula (IB), wherein:
  • R 1 is hydrogen
  • R 2 is a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
  • R 2 can not be /?-methylphenyl
  • R 3 is methyl, ethyl, isopropyl, or tert-butyl
  • R 4 is hydrogen
  • R 5 and R 6 are each hydrogen
  • X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR 7 ;
  • R 7 is H, Ci-C 5 alkyl, or phenyl, wherein each substituent group of R 7 is optionally independently substituted with one, two, or three substituent groups selected from C 1 -C 3 alkyl, C 1 -C 3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl;
  • R 8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
  • each substituent group of R 8 is independently C 1 -C 3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C 1 -C 3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, trifluoromethyl, or C 1 -C 3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
  • each substituent group of R 8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl,
  • Preferred compounds of Formula (IB) include the following:
  • More preferred compounds of Formula (IB) include the following:
  • the invention also provides a method of making a compound of Formula (IB)
  • R 1 , R 4 , R 5 , and R 6 are each ⁇ , and R 2 , R 3 , R 8 , and X are as defined above, the method comprising: (a) reacting the amino alcohol of Formula (XII) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as tetrahydrofuran, in the presence of a base, such as sodium hydride, or in dichloromethane in the presence of pyridine followed by a suitable base, such as aqueous potassium hydroxide, to form an aziridine of Formula (XIII)
  • XIII III IB or (c) reacting the aziridine of Formula (XIII) with an aniline or thiol (X is NR 7 or S) reagent R 8 -XH of Formula (XIV) in a suitable solvent, such as tetrahydrofuran, methanol, or acetonitrile, under thermal conditions with or without an additive such as lithium perchlorate, ⁇ -cyclodextrin hydrate, or triethylamine to form the compound of Formula (IB)
  • a suitable solvent such as tetrahydrofuran, methanol, or acetonitrile
  • the group R 2 may be substituted with another R 2 group, the method comprising:
  • the intermediate aziridine of Formula (XIII) may be made by the following method:
  • the intermediate aziridine of Formula (XIII) may be made by the following method:
  • the compounds according to the invention are formulated into pharmaceutical compositions comprising an effective amount, preferably a pharmaceutically effective amount, of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof, and a pharmaceutically acceptable excipient or carrier.
  • the invention also provides a method of modulating the glucocorticoid receptor function in a patient, the method comprising administering to the patient an effective amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
  • the invention further provides a method of treating a disease-state or condition mediated by the glucocorticoid receptor function in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
  • the invention also provides a method of treating a disease-state or condition selected from: type II diabetes, obesity, cardiovascular diseases, hypertension, arteriosclerosis, neurological diseases, adrenal and pituitary tumors, and glaucoma, in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
  • a disease-state or condition selected from: type II diabetes, obesity, cardiovascular diseases, hypertension, arteriosclerosis, neurological diseases, adrenal and pituitary tumors, and glaucoma
  • the invention provides a method of treating a disease characterized by inflammatory, allergic, or proliferative processes, in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
  • the disease characterized by inflammatory, allergic, or proliferative processes is selected from: (i) lung diseases; (ii) rheumatic diseases or autoimmune diseases or joint diseases; (iii) allergic diseases; (iv) vasculitis diseases; (v) dermatological diseases; (vi) renal diseases; (vii) hepatic diseases; (viii) gastrointestinal diseases; (ix) proctological diseases; (x) eye diseases; (xi) diseases of the ear, nose, and throat (ENT) area; (xii) neurological diseases; (xiii) blood diseases; (xiv) tumor diseases; (xv) endocrine diseases; (xvi) organ and tissue transplantations and graft-versus-host diseases; (xvii) severe states of shock; (xviii) substitution therapy; and (xix) pain of inflammatory genesis.
  • the disease characterized by inflammatory, allergic, or proliferative processes is selected from: type I diabetes, osteoarthritis, Guillain-Barre syndrome, restenosis following percutaneous transluminal coronary angioplasty, Alzheimer disease, acute and chronic pain, atherosclerosis, reperfusion injury, bone resorption diseases, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, acute purulent meningitis, necrotizing enterocolitis, and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion.
  • the invention further provides methods of treating the disease-states or conditions mentioned above, in a patient in need of such treatment, the methods comprising sequentially or simultaneously administering to the patient: (a) an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) a pharmaceutically acceptable glucocorticoid.
  • the invention further provides a method of assaying the glucocorticoid receptor function in a sample, comprising: (a) contacting the sample with a selected amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) detecting the amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof bound to glucocorticoid receptors in the sample.
  • the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof is labeled with a detectable marker selected from: a radiolabel, fluorescent tag, a chemiluminescent tag, a chromophore, and a spin label.
  • a detectable marker selected from: a radiolabel, fluorescent tag, a chemiluminescent tag, a chromophore, and a spin label.
  • the invention also provides a method of imaging the glucocorticoid receptor distribution in a sample or patient, the method comprising: (a) contacting the sample or administering to a patient a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker; (b) detecting the spatial distribution and amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker bound to glucocorticoid receptors in the sample or patient using an imaging means to obtain an image; and (c) displaying an image of the spatial distribution and amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker bound to glucocorticoid receptors in the sample.
  • the imaging means is selected from: radioscintigraphy, nuclear magnetic resonance imaging (MRI), computed tomography (CT scan), or positron emission tomography (PET).
  • MRI nuclear magnetic resonance imaging
  • CT scan computed tomography
  • PET positron emission tomography
  • the invention also provides a kit for the in vitro diagnostic determination of the glucocorticoid receptor function in a sample, comprising: (a) a diagnostically effective amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) instructions for use of the diagnostic kit.
  • Ci-Ci 0 alkyl means an alkyl group or radical having 1 to 10 carbon atoms.
  • the term "lower” applied to any carbon-containing group means a group containing from 1 to 8 carbon atoms, as appropriate to the group (i.e., a cyclic group must have at least 3 atoms to constitute a ring).
  • alkylaryl means a monovalent radical of the formula AIk-Ar-
  • arylalkyl means a monovalent radical of the formula Ar-AIk- (where AIk is an alkyl group and Ar is an aryl group).
  • use of a term designating a monovalent radical where a divalent radical is appropriate shall be construed to designate the respective divalent radical and vice versa.
  • conventional definitions of terms control and conventional stable atom valences are presumed and achieved in all formulas and groups.
  • alkyl or "alkyl group” mean a branched or straight-chain saturated aliphatic hydrocarbon monovalent radical. This term is exemplified by groups such as methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1,1 -dimethyl ethyl (tert-butyl), and the like. It may be abbreviated "AIk”.
  • alkenyl or “alkenyl group” mean a branched or straight-chain aliphatic hydrocarbon monovalent radical containing at least one carbon-carbon double bond.
  • This term is exemplified by groups such as ethenyl, propenyl, n-butenyl, isobutenyl, 3-methylbut- 2-enyl, n-pentenyl, heptenyl, octenyl, decenyl, and the like.
  • alkynyl or "alkynyl group” mean a branched or straight-chain aliphatic hydrocarbon monovalent radical containing at least one carbon-carbon triple bond. This term is exemplified by groups such as ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, n- pentynyl, heptynyl, octynyl, decynyl, and the like.
  • alkylene or "alkylene group” mean a branched or straight-chain saturated aliphatic hydrocarbon divalent radical having the specified number of carbon atoms. This term is exemplified by groups such as methylene, ethylene, propylene, n-butylene, and the like, and may alternatively and equivalently be denoted herein as -(alkyl)-.
  • alkenylene or "alkenylene group” mean a branched or straight-chain aliphatic hydrocarbon divalent radical having the specified number of carbon atoms and at least one carbon-carbon double bond. This term is exemplified by groups such as ethenylene, propenylene, n-butenylene, and the like, and may alternatively and equivalently be denoted herein as -(alkylenyl)-.
  • alkynylene or "alkynylene group” mean a branched or straight-chain aliphatic hydrocarbon divalent radical containing at least one carbon-carbon triple bond. This term is exemplified by groups such as ethynylene, propynylene, n-butynylene, 2-butynylene, 3- methylbutynylene, n-pentynylene, heptynylene, octynylene, decynylene, and the like, and may alternatively and equivalently be denoted herein as -(alkynyl)-.
  • alkoxy or "alkoxy group” mean a monovalent radical of the formula AIkO-, where AIk is an alkyl group. This term is exemplified by groups such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, sec-butoxy, tert-butoxy, pentoxy, and the like.
  • aryloxy means a monovalent radical of the formula ArO-, where Ar is aryl. This term is exemplified by groups such as phenoxy, naphthoxy, and the like.
  • alkylcarbonyl means a monovalent radical of the formula AIkC(O)-, where AIk is alkyl or hydrogen.
  • arylcarbonyl means a monovalent radical of the formula ArC(O)-, where Ar is aryl.
  • acyl or "acyl group” mean a monovalent radical of the formula RC(O)-, where R is a substituent selected from hydrogen or an organic substituent.
  • substituents include alkyl, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, and the like. As such, the terms comprise alkylcarbonyl groups and arylcarbonyl groups.
  • acylamino or "acylamino group” mean a monovalent radical of the formula RC(O)N(R)-, where each R is a substituent selected from hydrogen or a substituent group.
  • alkoxycarbonyl or "alkoxycarbonyl group” mean a monovalent radical of the formula AIkO-C(O)-, where AIk is alkyl.
  • exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, tert-butyloxycarbonyl, and the like.
  • aryloxycarbonyl or "aryloxycarbonyl group” mean a monovalent radical of the formula ArO-C(O)-, where Ar is aryl.
  • alkylcarbonyloxy or “alkylcarbonyloxy group” or “alkanoyloxy” or “alkanoyloxy group” mean a monovalent radical of the formula AIkC(O)O-, where AIk is alkyl.
  • arylcarbonyloxy or “arylcarbonyloxy group” or “aroyloxy” or “aroyloxy group” mean a monovalent radical of the formula ArC(O)O-, where Ar is aryl.
  • alkylaminocarbonyloxy or “alkylaminocarbonyloxy group” mean a monovalent radical of the formula R 2 NC(O)O-, where each R is independently hydrogen or lower alkyl.
  • alkoxycarbonylamino or "alkoxycarbonylamino group” mean a monovalent radical of the formula ROC(O)NH-, where R is lower alkyl.
  • alkylcarbonylamino or “alkylcarbonylamino group” or “alkanoylamino” or “alkanoylamino groups” mean a monovalent radical of the formula AIkC(O)NH-, where AIk is alkyl.
  • exemplary alkylcarbonylamino groups include acetamido (CHsC(O)NH-).
  • alkylaminocarbonyloxy or “alkylaminocarbonyloxy group” mean a monovalent radical of the formula AIkNHC(O)O-, where AIk is alkyl.
  • amino or “amino group” mean an -NH 2 group.
  • alkylamino or "alkylamino group” mean a monovalent radical of the formula (AIk)NH-, where AIk is alkyl.
  • exemplary alkylamino groups include methylamino, ethylamino, propylamino, butylamino, tert-butylamino, and the like.
  • dialkylamino or "dialkylamino group” mean a monovalent radical of the formula (AIk)(AIk)N-, where each AIk is independently alkyl.
  • exemplary dialkylamino groups include dimethylamino, methylethylamino, diethylamino, dipropylamino, ethylpropylamino, and the like.
  • substituted amino or “substituted amino group” mean a monovalent radical of the formula -NR 2 , where each R is independently a substituent selected from hydrogen or the specified substituents (but where both Rs cannot be hydrogen).
  • substituents include alkyl, alkanoyl, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, and the like.
  • alkoxycarbonylamino or “alkoxycarbonylamino group” mean a monovalent radical of the formula AIkOC(O)NH-, where AIk is alkyl.
  • ureido or “ureido group” mean a monovalent radical of the formula R. 2 NC(O)NH-, where each R is independently hydrogen or alkyl.
  • halo means one or more hydrogen atoms of the group are replaced by halogen groups.
  • haloalkyl or "haloalkyl group” mean a branched or straight-chain saturated aliphatic hydrocarbon monovalent radical, wherein one or more hydrogen atoms thereof are each independently replaced with halogen atoms. This term is exemplified by groups such as chloromethyl, 1 ,2-dibromoethyl, 1 , 1 , 1 -trifluoropropyl, 2-iodobutyl, l-chloro-2-bromo-3- fluoropentyl, and the like.
  • sulfanyl means a divalent radical of the formula -S-.
  • alkylthio or "alkylthio group” mean a monovalent radical of the formula AIkS-, where AIk is alkyl.
  • exemplary groups include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, and the like.
  • arylthio or "arylthio group” mean a monovalent radical of the formula ArS-, where Ar is aryl.
  • sulfinyl means a divalent radical of the formula -SO-.
  • sulfonyl or "sulfonyl group” mean a divalent radical of the formula -SO 2 -.
  • sulfonylamino or “sulfonylamino group” mean a divalent radical of the formula -SO 2 NR-, where R is a hydrogen or a substituent group.
  • aminonosulfonyl or “aminosulfonyl group” mean a monovalent radical of the formula NR 2 SO 2 -, where R is each independently a hydrogen or a substituent group.
  • carbocycle or “carbocyclic group” mean a stable aliphatic 3- to 15-membered monocyclic or polycyclic monovalent or divalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the carbocycle may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • the term comprises cycloalkyl (including spiro cycloalkyl), cycloalkylene, cycloalkenyl, cycloalkenylene, cycloalkynyl, and cycloalkynylene, and the like.
  • cycloalkyl or "cycloalkyl group” mean a stable aliphatic saturated 3- to 15- membered monocyclic or polycyclic monovalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornane, adamantyl, tetrahydronaphthyl (tetralin), 1-decalinyl, bicyclo[2.2.2]octanyl, 1 -methylcyclopropyl, 2-methylcyclopentyl, 2- methylcyclooctyl, and the like.
  • cycloalkenyl or “cycloalkenyl group” mean a stable aliphatic 5- to 15-membered monocyclic or polycyclic monovalent radical having at least one carbon-carbon double bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring.
  • the cycloalkenyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl, cyclodecenyl, norbornenyl, 2-methylcyclopentenyl, 2-methylcyclooctenyl, and the like.
  • cycloalkynyl or “cycloalkynyl group” mean a stable aliphatic 8- to 15-membered monocyclic or polycyclic monovalent radical having at least one carbon-carbon triple bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 8- to 10-membered monocyclic or 12- to 15-membered bicyclic ring.
  • the cycloalkynyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkynyl groups include, cyclooctynyl, cyclononynyl, cyclodecynyl, 2-methylcyclooctynyl, and the like.
  • cycloalkylene or "cycloalkylene group” mean a stable saturated aliphatic 3- to 15-membered monocyclic or polycyclic divalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkylene groups include cyclopentylene, and the like.
  • cycloalkenylene or "cycloalkenylene group” mean a stable aliphatic 5- to 15- membered monocyclic or polycyclic divalent radical having at least one carbon-carbon double bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring.
  • the cycloalkenylene ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkenylene groups include cyclopentenylene, cyclohexenylene, cycloheptenylene, cyclooctenylene, cyclononenylene, cyclodecenylene, norbornenylene, 2-methylcyclopentenylene, 2- methylcyclooctenylene, and the like.
  • cycloalkynylene or "cycloalkynylene group” mean a stable aliphatic 8- to 15- membered monocyclic or polycyclic divalent radical having at least one carbon-carbon triple bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 8- to 10-membered monocyclic or 12- to 15-membered bicyclic ring. Unless otherwise specified, the cycloalkynylene ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • Exemplary cycloalkynylene groups include cyclooctynylene, cyclononynylene, cyclodecynylene, 2-methylcyclooctynylene, and the like.
  • aryl or “aryl group” mean an aromatic carbocyclic monovalent or divalent radical of from 6 to 14 carbon atoms having a single ring (e.g., phenyl or phenylene) or multiple condensed rings (e.g., naphthyl or anthranyl). Unless otherwise specified, the aryl ring may be attached at any suitable carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure.
  • aryl groups include phenyl, naphthyl, dihydronaphthyl, tetrahydronaphthyl, anthryl, phenanthryl, indanyl, indenyl, biphenyl, and the like. It may be abbreviated "Ar”.
  • heteroaryl or “heteroaryl group” mean a stable aromatic 5- to 14-membered, monocyclic or polycyclic monovalent or divalent radical which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic radical, having from one to four heteroatoms in the ring(s) independently selected from nitrogen, oxygen, and sulfur, wherein any sulfur heteroatoms may optionally be oxidized and any nitrogen heteroatom may optionally be oxidized or be quaternized.
  • the heteroaryl ring may be attached at any suitable heteroatom or carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable heteroatom or carbon atom which results in a stable structure.
  • exemplary and preferred heteroaryls include furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, indolyl, azaindolyl, dihydroindolyl, isoindolyl, benzofuranyl, dihydrobenzofuranyl, benzopyranyl, dihydrobenzopyranyl, benzpyrrolyl,
  • heterocycle means a stable non-aromatic 5- to 14-membered monocyclic or polycyclic, monovalent or divalent, ring which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring, having from one to three heteroatoms in the ring(s) independently selected from nitrogen, oxygen, and sulfur, wherein any sulfur heteroatoms may optionally be oxidized and any nitrogen heteroatom may optionally be oxidized or be quaternized.
  • the heterocyclyl ring may be attached at any suitable heteroatom or carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable heteroatom or carbon atom which results in a stable structure.
  • exemplary and preferred heterocycles include pyrrolinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrofuranyl, hexahydropyrimidinyl, hexahydropyridazinyl, and the like.
  • compounds of the invention and equivalent expressions are meant to embrace compounds of Formula (I) as herein described, including the tautomers, the prodrugs, the salts, particularly the pharmaceutically acceptable salts, and the solvates and hydrates thereof, where the context so permits.
  • the compounds of the invention and the formulas designating the compounds of the invention are understood to only include the stable compounds thereof and exclude unstable compounds, even if an unstable compound might be considered to be literally embraced by the compound formula.
  • reference to intermediates, whether or not they themselves are claimed is meant to embrace their salts and solvates, where the context so permits.
  • stable compound or “stable structure” mean a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic or diagnostic agent.
  • a compound which would have a "dangling valency" or is a carbanion is not a compound contemplated by the invention.
  • substituted means that any one or more hydrogens on an atom of a group or moiety, whether specifically designated or not, is replaced with a selection from the indicated group of substituents, provided that the atom's normal valency is not exceeded and that the substitution results in a stable compound. If a bond to a substituent is shown to cross the bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound, then such substituent may be bonded via any atom in such substituent.
  • such piperazinyl, piperidinyl, or tetrazolyl group may be bonded to the rest of the compound of the invention via any atom in such piperazinyl, piperidinyl, or tetrazolyl group.
  • any substituent or group occurs more than one time in any constituent or compound, its definition on each occurrence is independent of its definition at every other occurrence.
  • the term “about” or “approximately” means within 20%, preferably within 10%, and more preferably within 5% of a given value or range.
  • the yield of each of the reactions described herein is expressed as a percentage of the theoretical yield.
  • prodrug or “prodrug derivative” mean a covalently-bonded derivative or carrier of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s).
  • prodrugs have metabolically cleavable groups and are rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood, and generally include esters and amide analogs of the parent compounds.
  • the prodrug is formulated with the objectives of improved chemical stability, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity).
  • prodrugs themselves have weak or no biological activity and are stable under ordinary conditions.
  • Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: "Design and Applications of Prodrugs”; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery. K.B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzymology, K. Widder et al. (eds.), Vol. 42, Academic Press, 1985, particularly pp.
  • pharmaceutically acceptable prodrug means a prodrug of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible.
  • salt means an ionic form of the parent compound or the product of the reaction between the parent compound with a suitable acid or base to make the acid salt or base salt of the parent compound. Salts of the compounds of the present invention can be synthesized from the parent compounds which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts are prepared by reacting the free base or acid parent compound with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base in a suitable solvent or various combinations of solvents.
  • pharmaceutically acceptable salt means a salt of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use.
  • pharmaceutically- acceptable acid addition salts and pharmaceutically-acceptable base addition salts.
  • the compounds of the present invention are useful in both free base and salt form, in practice, the use of the salt form amounts to use of the base form. Lists of suitable salts are found in, e.g., S.M. Birge et at, J. Pharm. ScL, 1977, 66, pp. 1-19, which is hereby incorporated by reference in its entirety.
  • pharmaceutically-acceptable acid addition salt means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trichloroacetic acid, trifluoroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 2- acetoxybenzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, hemisulfic acid, heptanoic acid, hexanoic acid, formic acid,
  • pharmaceutically-acceptable base addition salt means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a metal cation such as sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically-acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, quaternary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion-exchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2- dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tetramethylammonium compounds, tetraethylammonium compounds
  • solvate means a physical association of a compound with one or more solvent molecules or a complex of variable stoichiometry formed by a solute (for example, a compound of Formula (I)) and a solvent, for example, water, ethanol, or acetic acid. This physical association may involve varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. In general, the solvents selected do not interfere with the biological activity of the solute. Solvates encompasses both solution-phase and isolatable solvates. Representative solvates include hydrates, ethanolates, methanolates, and the like.
  • hydrate means a solvate wherein the solvent molecule(s) is/are H 2 O.
  • the compounds of the present invention as discussed below include the free base or acid thereof, their salts, solvates, and prodrugs and may include oxidized sulfur atoms or quaternized nitrogen atoms in their structure, although not explicitly stated or shown, particularly the pharmaceutically acceptable forms thereof. Such forms, particularly the pharmaceutically acceptable forms, are intended to be embraced by the appended claims.
  • isomers means compounds having the same number and kind of atoms, and hence the same molecular weight, but differing with respect to the arrangement or configuration of their atoms in space.
  • the term includes stereoisomers and geometric isomers.
  • stereoisomer or “optical isomer” means a stable isomer that has at least one chiral atom or restricted rotation giving rise to perpendicular dissymmetric planes (e.g., certain biphenyls, allenes, and spiro compounds) and can rotate plane-polarized light. Because asymmetric centers and other chemical structure exist in the compounds of the invention which may give rise to stereoisomerism, the invention contemplates stereoisomers and mixtures thereof.
  • the compounds of the invention and their salts include asymmetric carbon atoms and may therefore exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers. Typically, such compounds will be prepared as a racemic mixture.
  • stereoisomers can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures.
  • individual stereoisomers of compounds are prepared by synthesis from optically active starting materials containing the desired chiral centers or by preparation of mixtures of enantiomeric products followed by separation or resolution, such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, use of chiral resolving agents, or direct separation of the enantiomers on chiral chromatographic columns.
  • Starting compounds of particular stereochemistry are either commercially available or are made by the methods described below and resolved by techniques well-known in the art.
  • enantiomers means a pair of stereoisomers that are non-superimposable mirror images of each other.
  • diastereoisomers or “diastereomers” mean stereoisomers which are not mirror images of each other.
  • racemic mixture or “racemate” mean a mixture containing equal parts of individual enantiomers.
  • non-racemic mixture means a mixture containing unequal parts of individual enantiomers.
  • Determination of the E and Z isomers can be done by analytical methods such as x-ray crystallography, 1 H NMR, and 13 C NMR. Some of the compounds of the invention can exist in more than one tautomeric form. As mentioned above, the compounds of the invention include all such tautomers.
  • enantiomers often exhibit strikingly different biological activity including differences in pharmacokinetic properties, including metabolism, protein binding, and the like, and pharmacological properties, including the type of activity displayed, the degree of activity, toxicity, and the like.
  • one enantiomer may be more active or may exhibit beneficial effects when enriched relative to the other enantiomer or when separated from the other enantiomer.
  • one skilled in the art would know how to separate, enrich, or selectively prepare the enantiomers of the compounds of the invention from this disclosure and the knowledge of the prior art.
  • racemic form of drug may be used, it is often less effective than administering an equal amount of enantiomerically pure drug; indeed, in some cases, one enantiomer may be pharmacologically inactive and would merely serve as a simple diluent.
  • ibuprofen had been previously administered as a racemate, it has been shown that only the 5 * -isomer of ibuprofen is effective as an anti-inflammatory agent (in the case of ibuprofen, however, although the R -isomer is inactive, it is converted in vivo to the S- isomer, thus, the rapidity of action of the racemic form of the drug is less than that of the pure .S-isomer).
  • enantiomers may have distinct biological activity.
  • ⁇ -penicillamine is a therapeutic agent for chronic arthritis, while ⁇ -penicillamine is toxic.
  • some purified enantiomers have advantages over the racemates, as it has been reported that purified individual isomers have faster transdermal penetration rates compared to the racemic mixture. See U.S. Patent Nos. 5,114,946 and 4,818,541.
  • one enantiomer is pharmacologically more active, less toxic, or has a preferred disposition in the body than the other enantiomer, it would be therapeutically more beneficial to administer that enantiomer preferentially. In this way, the patient undergoing treatment would be exposed to a lower total dose of the drug and to a lower dose of an enantiomer that is possibly toxic or an inhibitor of the other enantiomer.
  • Preparation of pure enantiomers or mixtures of desired enantiomeric excess (ee) or enantiomeric purity are accomplished by one or more of the many methods of (a) separation or resolution of enantiomers, or (b) enantioselective synthesis known to those of skill in the art, or a combination thereof.
  • These resolution methods generally rely on chiral recognition and include, for example, chromatography using chiral stationary phases, enantioselective host-guest complexation, resolution or synthesis using chiral auxiliaries, enantioselective synthesis, enzymatic and nonenzymatic kinetic resolution, or spontaneous enantioselective crystallization.
  • patient includes both human and non-human mammals.
  • the term "effective amount” means an amount of a compound according to the invention which, in the context of which it is administered or used, is sufficient to achieve the desired effect or result. Depending on the context, the term effective amount may include or be synonymous with a pharmaceutically effective amount or a diagnostically effective amount.
  • pharmaceutically effective amount or “therapeutically effective amount” means an amount of a compound according to the invention which, when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue, system, or patient that is sought by a researcher or clinician.
  • the amount of a compound of according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the invention, and the age, body weight, general health, sex, and diet of the patient.
  • a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure.
  • diagnostically effective amount means an amount of a compound according to the invention which, when used in a diagnostic method, apparatus, or assay, is sufficient to achieve the desired diagnostic effect or the desired biological activity necessary for the diagnostic method, apparatus, or assay. Such an amount would be sufficient to elicit the biological or medical response in a diagnostic method, apparatus, or assay, which may include a biological or medical response in a patient or in a in vitro or in vivo tissue or system, that is sought by a researcher or clinician.
  • the amount of a compound according to the invention which constitutes a diagnostically effective amount will vary depending on such factors as the compound and its biological activity, the diagnostic method, apparatus, or assay used, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of administration, drugs and other compounds used in combination with or coincidentally with the compounds of the invention, and, if a patient is the subject of the diagnostic administration, the age, body weight, general health, sex, and diet of the patient.
  • a diagnostically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure.
  • modulate means the ability of a compound to alter the function of the glucocorticoid receptor by, for example, binding to and stimulating or inhibiting the glucocorticoid receptor functional responses.
  • modulator in the context of describing compounds according to the invention means a compound that modulates the glucocorticoid receptor function.
  • modulators include, but are not limited to, agonists, partial agonists, antagonists, and partial antagonists.
  • agonist in the context of describing compounds according to the invention means a compound that, when bound to the glucocorticoid receptor, enhances or increases the glucocorticoid receptor function.
  • agonists include partial agonists and full agonists.
  • full agonist in the context of describing compounds according to the invention means a compound that evokes the maximal stimulatory response from the glucocorticoid receptor, even when there are spare (unoccupied) glucocorticoid receptors present.
  • partial agonist in the context of describing compounds according to the invention means a compound that is unable to evoke the maximal stimulatory response from the glucocorticoid receptor, even at concentrations sufficient to saturate the glucocorticoid receptors present.
  • antagonist in the context of describing compounds according to the invention means a compound that directly or indirectly inhibits or suppresses the glucocorticoid receptor function.
  • antagonists include partial antagonists and full antagonists.
  • full antagonist in the context of describing compounds according to the invention means a compound that evokes the maximal inhibitory response from the glucocorticoid receptor, even when there are spare (unoccupied) glucocorticoid receptors present.
  • partial antagonist in the context of describing compounds according to the invention means a compound that is unable to evoke the maximal inhibitory response from the glucocorticoid receptor, even at concentrations sufficient to saturate the glucocorticoid receptors present.
  • treating or “treatment” mean the treatment of a disease-state in a patient, and include:
  • reaction conditions and reaction times may vary depending on the particular reactants used. Unless otherwise specified, solvents, temperatures, pressures, and other reaction conditions may be readily selected by one of ordinary skill in the art. Specific procedures are provided in the Synthetic Examples section. Typically, reaction progress may be monitored by thin layer chromatography (TLC), if desired, and intermediates and products may be purified by chromatography on silica gel and/or by recrystallization.
  • TLC thin layer chromatography
  • an amino acid where R' is H or an amino acid ester where R' is Me or Et is reacted with a sulfonyl chloride (V) in a suitable solvent, such as THF, in the presence of a suitable base, such as NaH, to provide sulfonamide (VI).
  • a suitable solvent such as THF
  • a suitable base such as NaH
  • Reduction of sulfonamide (VI) with a suitable reducing agent, such as lithium aluminum hydride provides alcohol (VII).
  • Ring closure by methods known in the art, for example, reacting the alcohol with a sulfonyl chloride such a /? ⁇ r ⁇ -toluenesulfonyl chloride in the presence of a suitable base, such as sodium hydride, provides the aziridine (II).
  • a sulfonyl chloride such as /? ⁇ r ⁇ -toluenesulfonyl chloride in the presence of a suitable base, such as sodium hydride
  • the aziridine (II) is reacted with a reagent of R 6 X-M of Formula (III) where X is sulfur, oxygen, or NR 7 and M is Na, K or Li derived from R 6 X-H were X is oxygen using a suitable base such as sodium hydride in a suitable solvent such as DMF or DMSO; and from R 6 X-H were X is sulfur using a suitable base such as sodium hydride in a suitable solvent such as DMF or DMSO, or 2-tert- butylimino-2-diehylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine on polystyrene in a suitable solvent such as acetonitrile and from R 6 X-H were X is NR 7 using sodium bis(trimethylsilyl)amide in a suitable solvent such a DMSO or DMF.
  • the aziridine (II) is reacted with a reagent R 6 X-M of Formula (III) where X is NR 7 or sulfur and M is hydrogen in a suitable solvent such as THF under thermal conditions to form the compound of Formula (IA).
  • a suitable solvent such as THF
  • the racemic and chiral amino acids and amino acid esters (IV) as well as the sulfonyl chlorides R SO 2 CI (V) are either commercially available or may be readily prepared by methods known to those skilled in the art.
  • enantiomerically enriched compounds of Formula (IA) may be prepared by using chiral starting materials. For example, a method of preparing 1 , 1 , 1 -trifluoroalanine is given in V.A. Soloshonok et ah, Tetrahedron, 1997, 53, 8307.
  • an amine of Formula (VIII) is reacted with a sulfonyl chloride of Formula (V) in a suitable solvent such as pyridine in to form an sulfonamide of Formula (IX).
  • a suitable solvent such as pyridine
  • an oxonium salt such as trimethyloxonium tetrafluoroborate
  • a suitable solvent such as dichloromethane
  • Chiral ⁇ -fluorinated amines of Formula (VIII) as well as the sulfonyl chlorides R 2 SC ⁇ Cl (V) are either commercially available or may be readily prepared by methods known to those skilled in the art.
  • enantiomerically enriched compounds of Formula (IA) may be prepared by using chiral starting materials.
  • a method of preparing chiral and racemic ⁇ -fluorinated amines of Formula (VIII) is given in P. Bravo et ah, J. Org. Chem., 1996, 61, 3375.
  • a method of preparing chiral aziridines is given in A. Toshimitsu et ah, J. Chem. Soc, Chem. Commun, 1992, 284.
  • Compounds of Formula (IB) may be prepared using a general procedure outlined in Scheme III. This general procedure is suitable for a variety of R 2 and R 3 (for this example, R 3 is ethyl) where R 8 is an optionally substituted heteroaryl group and X is sulfur, oxygen, or nitrogen (for this example, X is nitrogen and R 7 is H).
  • the optionally substituted amino alcohol (XII) bearing R 3 is reacted with a sulfonyl chloride (V) bearing R in a suitable solvent such as tetrahydrofuran in the presence of a base such as sodium hydride or in dichloromethane in the presence of pyridine followed cyclization with a suitable base such as aqueous potassium hydroxide to form an aziridine of Formula (XIII).
  • a suitable solvent such as tetrahydrofuran in the presence of a base such as sodium hydride or in dichloromethane in the presence of pyridine
  • a suitable base such as aqueous potassium hydroxide
  • the aziridine (XIII) is reacted with a suitable organometallic reagent R 8 X-M where X is sulfur, oxygen, or NR 7 and M is Li, Na, or K derived from R 8 X-H were X is oxygen using a suitable base such as sodium hydride in a suitable solvent, such as DMF or DMSO; and from R 8 X-H were X is sulfur using a suitable base such as sodium hydride in a suitable solvent, such as DMF or DMSO, or 2-tert- butylimino-2-diethylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine on polystyrene in a suitable solvent, such as acetonitrile, and from R 8 X-H where X is NR 7 using sodium bis(trimethylsilyl)amide in a suitable solvent, such a DMSO or DMF.
  • R 8 X-M where X is sulfur, oxygen, or NR 7 and M
  • the aziridine (XIII) is reacted with a reagent R 8 X-M where X is NR 7 or sulfur and M is hydrogen in a suitable solvent, such as THF, under thermal conditions with or without an additive, such as lithium perchlorate in acetonitrile or ⁇ -cyclodextrin hydrate in methanol, to form the compound of Formula (IB).
  • a suitable solvent such as THF
  • Racemic and chiral aminoalcohols are either commercially available or may be readily prepared by methods known to those skilled in the art.
  • the sulfonyl chlorides R 2 SC ⁇ Cl (V) are either commercially available or may be readily prepared by methods known to those skilled in the art.
  • a general method of preparing sulfonyl chloride from anilines is given in R.V. Hoffman, Org. Synth. 1981, 60, 121.
  • Aziridines (XIII) may also be commercially available or prepared from aminoalcohols by methods known to those skilled in the art. For example, methods of preparing aziridines are given in M.B. Berry and D. Craig, Synlett 1992, 41; J. Farras, et al.
  • a compound of Formula (IB) bearing R 8 , bearing R 3 (for this example, R 3 is ethyl), bearing XR 7 (for this example, X is nitrogen and R 7 is H), and an R 2 (for this example, R 2 is o-nitropheny) is reacted with thiophenol and a suitable base such as potassium carbonate in a suitable solvent such as DMF to provide the amine (XX).
  • the aminoethyl compound of Formula (XX) is sulfonated with a sulfonyl halide (for this example, R 2 is 2,4-dichloro-6-aminophenyl) of Formula (XXI) in the presence of a suitable base such as triethyamine or pyridine in a suitable solvent such as dichloromethane to form the compound of Formula (IB) where R is not o-nitrophenyl.
  • a suitable base such as triethyamine or pyridine
  • a suitable solvent such as dichloromethane
  • the optionally substituted amino acid of Formula (XXII) useful to provide compounds of Formula (IB) bearing R 3 is reacted with a sulfonyl chloride (VII) in a suitable solvent, such as acetone-water, in the presence of a suitable base, such as NaOH to provide sulfonamide of Formula (XXIII).
  • a suitable solvent such as acetone-water
  • a suitable base such as NaOH
  • an activating agent such as thiophosgene
  • a suitable solvent such as chloroform
  • Reduction of an amide of Formula (XXV) with a suitable reducing agent, such as borane in a suitable solvent, such as THF provides compounds of Formula (IB).
  • compounds of (IA) can be obtained from 1 ,1 ,1-trifluoroalanine.
  • An example of such a probe is rhodamine (TAMRA)-labeled dexamethasone.
  • the affinity of the probe for the steroid receptor was then determined in a titration experiment.
  • the fluorescence polarization value of the probe in assay buffer was measured on an SLM-8100 fluorometer using the excitation and emission maximum values described above. Aliquots of expression vector lysate were added and fluorescence polarization was measured after each addition until no further change in polarization value was observed. Non-linear least squares regression analysis was used to calculate the dissociation constant of the probe from the polarization values obtained for lysate binding to the probe.
  • Step 2 Screening for Inhibitors of Probe Binding
  • FP fluorescence polarization
  • TAMRA tetramethylrhodamine
  • GR human glucocorticoid receptor
  • the assay buffer was: 10 mM TES, 50 mM KCl, 20 mM Na 2 MoO 4 » 2H 2 O, 1.5 mM EDTA, 0.04% w/v CHAPS, 10% v/v glycerol, 1 mM dithiothreitol, pH 7.4.
  • Test compounds were dissolved to 1 mM in neat DMSO and then further diluted to 1 Ox assay concentration in assay buffer supplemented with 10% v/v DMSO. Test compounds were serially diluted at 1Ox assay concentrations in 10% DMSO-containing buffer in 96-well polypropylene plates. Binding reaction mixtures were prepared in 96-well black Dynex microtiter plates by sequential addition of the following assay components to each well: 15 ⁇ L of 10x test compound solution, 85 ⁇ L of GR-containing baculovirus lysate diluted 1 : 170 in assay buffer, and 50 ⁇ L of 15 nM TAMRA-labeled dexamethasone.
  • Positive controls were reaction mixtures containing no test compound; negative controls (blanks) were reaction mixtures containing 0.7 ⁇ M to 2 ⁇ M dexamethasone.
  • the binding reactions were incubated for 1 hour at room temperature and then read for fluorescence polarization in the LJL Analyst set to 550 nm excitation and 580 nm emission, with the Rhodamine 561 dichroic mirror installed.
  • IC50 values were determined by iterative non-linear curve fitting of the FP signal data to a 4- parameter logistic equation.
  • PR insect cell lysate is diluted 1 :7.1 and MR lysate diluted 1 :9.4.
  • PR probe is TAMRA-labeled mifepristone, used at a final concentration of 5 nM in the assay, and the negative controls (blanks) were reactions containing mifepristone at 0.7 ⁇ M to 2 ⁇ M.
  • the ER protocol is similar to the above protocols, but uses PanVera kit receptor, fluorescein- labeled probe.
  • the assay components are made in the same volumes as above, to produce final assay concentrations for ER of 15 nM and ES2 probe of 1 nM.
  • the component order of addition is modified from the above assays: probe is added to the plate first, followed by receptor and test compound.
  • the plates are read in the LJL Analyst set to 485 nm excitation and 530 nm emission, with the Fluorescein 505 dichroic mirror installed.
  • Compounds found to bind to the glucocorticoid receptor may be evaluated for dissociation of transactivation and transrepression by assays cited in the Background of the Invention (CM. Bamberger and H.M. Schulte, Eur. J. Clin. Invest., 2000, 30 (suppl. 3) 6-9) or by the assays described below.
  • Dexamethasone a synthetic ligand to the glucocorticoid receptor (GR) induces expression of aromatase in human foreskin fibroblast cells.
  • the activity of aromatase is measured by the conversion of testosterone to estradiol in culture media.
  • Compounds that exhibit binding to GR are evaluated for their ability to induce aromatase activity in human foreskin fibroblasts.
  • Human foreskin fibroblast cells (ATCC Cat. No. CRL-2429, designation CCDl 12SK) are plated on 96 well plates at 50,000 cells per well 5 days before use, in Iscove's Modified Dulbecco's Media (GibcoBRL Life Technologies Cat No. 12440-053) supplemented with 10% charcoal filtered FBS (Clonetech Cat No. SH30068) and Gentamycin (GibcoBRL Life Technologies Cat. No. 15710-064). On the day of the experiment, the media in the wells is replaced with fresh media. Cells are treated with test compounds to final concentrations of 10 "5 M to 10 "8 M, and testosterone to a final concentration of 300 ng/mL.
  • Control wells include: (a) wells that receive testosterone only, and (b) wells that receive testosterone plus 2 ⁇ M of dexamethasone to provide maximum induction of aromatase. Plates are incubated at 37°C overnight (15 to 18 hours), and supernatants are harvested at the end of incubation. Estradiol in the supernatant is measured using ELISA kits for estradiol (made by ALPCO, obtained from American Laboratory Products Cat. No. 020-DR-2693) according to the manufacture's instruction. The amount of estradiol is inversely proportional to the ELISA signals in each well.
  • test compounds The extent of aromatase induction by test compounds is expressed as a relative percentage to dexamethasone. EC50 values of test compounds are derived by non- linear curve fitting. 2. Inhibition of IL-6 Production in Fibroblasts (Cell Assay for Transrepression)
  • Human foreskin fibroblast cells produce IL-6 in response to stimulation by pro-inflammatory cytokine IL-I .
  • This inflammatory response as measured by the production of IL-6, can be effectively inhibited by dexamethasone, a synthetic ligand to the glucocorticoid receptor (GR).
  • GR glucocorticoid receptor
  • Human foreskin fibroblast cells (ATCC Cat. No. CRL-2429) are plated on 96 well plates at 5,000 cells per well the day before use, in Iscove's Modified Dulbecco's Media (GibcoBRL Life Technologies Cat. No. 12440-053) supplemented with 10% charcoal filtered FBS (Clonetech Cat. No. SH30068) and Gentamycin (GibcoBRL Life Technologies Cat. No. 15710-064). On the next day, media in the wells is replaced with fresh media. Cells are treated with IL-I (rhIL-l ⁇ , R&D Systems Cat. No.
  • test compounds 200-LA to a final concentration of 1 ng/mL, and with test compounds to final concentrations of 10 ⁇ 5 M to 10 8 M, in a total volume of 200 ⁇ L per well.
  • Samples are done in duplicates. Background control wells do not receive test compounds or IL- 1.
  • Positive control wells receive IL- 1 only and represent maximum (or 100%) amount of IL-6 production. Plates are incubated at 37°C overnight (15 to 18 hours), and supernatants are harvested at the end of incubation. IL-6 levels in the supernatants are determined by the ELISA kits for IL-6 (MedSystems Diagnostics GmbH, Vienna, Austria, Cat. No. BMS213TEN) according to manufacture's instructions. The extent of inhibition of IL-6 by test compounds is expressed in percentage relative to positive controls. IC50 values of test compounds are derived by non-linear curve fitting.
  • Evaluation of agonist or antagonist activity of compounds binding to the glucocorticoid receptor may be determined by any of the assays.
  • TAT Tyrosine Aminotransferase
  • H4-II-E-C3 cells were incubated overnight in 96 well plates (20,000 cells/100 ⁇ L/well) in MEM medium containing 10% heat inactivated FBS and 1% nonessential amino acids. On the next day, cells were stimulated with the indicated concentrations of dexamethasone or test compound (dissolved in DMSO, final DMSO concentration 0.2%) for 18 hours. Control cells were treated with 0.2% DMSO. After 18 hours, the cells were lysed in a buffer containing 0.1% Triton X-IOO and the TAT activity was measured in a photometric assay using tyrosine and alpha-ketoglutarate as substrates.
  • the hepatoma cells were pre-stimulated by addition of dexamethasone (concentration ranges from 3 x 10 "9 M to 3 x 10 "8 M) shortly before the test compound was applied to the cells.
  • dexamethasone concentration ranges from 3 x 10 "9 M to 3 x 10 "8 M
  • the steroidal non-selective GR/PR antagonist mifepristone was used as control.
  • HeLa cells were stably co-transfected with the pHHLuc-plasmid containing a fragment of the MMTV-LTR (-200 to +100 relative to the transcription start site) cloned in front of the luciferase gene (Norden, 1988) and the pcDNA3.1 plasmid (Invitrogen) constitutively expressing the resistance for the selective antibiotic GENETICIN®. Clones with best induction of the MMTV-promoter were selected and used for further experiments.
  • the MMTV-promoter was pre-stimulated by adding dexamethasone (3 x 10 9 M to 3 x 10 8 M) shortly before the test compound was applied to the cells.
  • dexamethasone 3 x 10 9 M to 3 x 10 8 M
  • the steroidal non-selective GR/PR antagonist mifepristone was used as control. 5. Modulation of IL-8 Production in U937 Cells
  • U-937 cells were incubated for 2 to 4 days in RPMI1640 medium containing 10% CCS (charcoal treated calf serum). The cells were transferred to 96 well plates (40,000 cells/100 ⁇ L/well) and stimulated with 1 ⁇ g/mL LPS (dissolved in PBS) in the presence or absence of dexamethasone or test compound (dissolved in DMSO, final concentration 0.2%). Control cells were treated with 0.2% DMSO. After 18 hours, the IL-8 concentration in the cell supernatant was measured by ELISA, using the "OptEIA human IL-8 set" (Pharmingen, Cat. No. 2654KI).
  • the LPS-induced IL-8 secretion was inhibited by adding dexamethasone (3 x 10 "9 M to 3 x 10 "8 M) shortly before the test compound was applied to the cells.
  • dexamethasone 3 x 10 "9 M to 3 x 10 "8 M
  • the steroidal non-selective GR/PR antagonist mifepristone was used as control.
  • HeLa cells were stably co-transfected with a plasmid containing a 1.3 kb fragment of the human ICAM-promoter (-1353 to -9 relative to the transcription start site, Ledebur and Parks, 1995) cloned in front of the luciferase gene and the pcDNA3.1 plasmid (Invitrogen) which constitutively expresses the resistance for the antibiotic GENETICIN®.
  • Clones with best induction of the ICAM-promoter were selected and used for further experiments. Cells were transferred to 96 well plates (15,000 cells/ 100 ⁇ L/well) in DMEM medium supplemented with 3% CCS.
  • the activation of the ICAM-promoter was induced by addition of 10 ng/mL recombinant TNF-alpha (R&D System, Cat. No. 210-TA). Simultaneously the cells were treated with the test compound or dexamethasone (dissolved in DMSO, final concentration 0.2%). Control cells were treated with DMSO only. After 18 hours, the cells were lysed with cell lysis reagent (Promega, Cat. No. E1531), luciferase assay reagent (Promega, Cat. No. E1501) was added and glow luminescence was measured using a lumino meter (BMG, Offenburg).
  • the TNF-alpha-induced activation of the ICAM-promoter was inhibited by adding dexamethasone (3 x 10 ⁇ 9 M to 3 x 10 ⁇ 8 M) shortly before the test compound was applied to the cells.
  • dexamethasone 3 x 10 ⁇ 9 M to 3 x 10 ⁇ 8 M
  • the steroidal non-selective GR/PR antagonist mifepristone was used as control.
  • the preferred potency range (IC50) in the above assays is between 0.1 nM and 10 ⁇ M, the more preferred potency range is 0.1 nM to 1 ⁇ M, and the most preferred potency range is 0.1 nM to 100 nM.
  • Representative compounds of the invention have been tested and have shown activity as modulators of the glucocorticoid receptor function in one or more of the above assays.
  • MG-63 Human osteosarcoma MG-63 cells (ATCC, Cat. No. CRL-1427) are plated on 96 well plates at 20,000 cells per well the day before use in 200 ⁇ L media of 99% D-MEM/F-12 (Gibco- Invitrogen, Cat. No. 11039-021), supplemented with 1% penicillin and streptomycin (Gibco- Invitrogen, Cat. No. 15140-122), 10 ⁇ g/mL Vitamin C (Sigma, Cat. No. A-4544), and 1% charcoal filtered Fetal Bovine Serum (HyClone, Cat. No. SH30068.02).
  • the invention also provides methods of modulating the glucocorticoid receptor function in a patient comprising administering to the patient a compound according to the invention. If the purpose of modulating the glucocorticoid receptor function in a patient is to treat a disease- state or condition, the administration preferably comprises a therapeutically or pharmaceutically effective amount of a pharmaceutically acceptable compound according to the invention.
  • the administration preferably comprises an effective amount of a compound according to the invention, that is, the amount necessary to obtain the desired effect or degree of modulation.
  • the compounds of the invention are useful in modulating the glucocorticoid receptor function.
  • these compounds have therapeutic use in treating disease-states and conditions mediated by the glucocorticoid receptor function or that would benefit from modulation of the glucocorticoid receptor function.
  • the compounds of the invention modulate the glucocorticoid receptor function, they have very useful anti-inflammatory and antiallergic, immune-suppressive, and anti-proliferative activity and they can be used in patients as drugs, particularly in the form of pharmaceutical compositions as set forth below, for the treatment of disease-states and conditions.
  • the agonist compounds according to the invention can be used in patients as drugs for the treatment of the following disease-states or indications that are accompanied by inflammatory, allergic, and/or proliferative processes: (i) Lung diseases: chronic, obstructive lung diseases of any genesis, particularly bronchial asthma and chronic obstructive pulmonary disease (COPD); adult respiratory distress syndrome (ARDS); bronchiectasis; bronchitis of various genesis; all forms of restrictive lung diseases, particularly allergic alveolitis; all forms of lung edema, particularly toxic lung edema; all forms of interstitial lung diseases of any genesis, e.g., radiation pneumonitis; and sarcoidosis and granulomatoses, particularly
  • Lung diseases chronic, obstructive lung diseases of any genesis, particularly bronchial asthma and chronic obstructive pulmonary disease (COPD); adult respiratory distress syndrome (ARDS); bronchiectasis; bronchitis of various genesis; all forms
  • Rheumatic diseases or autoimmune diseases or joint diseases all forms of rheumatic diseases, especially rheumatoid arthritis, acute rheumatic fever, and polymyalgia rheumatica; reactive arthritis; rheumatic soft tissue diseases; inflammatory soft tissue diseases of other genesis; arthritic symptoms in degenerative joint diseases
  • Allergic diseases all forms of allergic reactions, e.g., angioneurotic edema, hay fever, insect bites, allergic reactions to drugs, blood derivatives, contrast agents, etc., anaphylactic shock (anaphylaxis), urticaria, angioneurotic edema, and contact dermatitis;
  • Vasculitis diseases panarteritis nodosa, polyarteritis nodosa, arteritis temporalis,
  • Dermatological diseases atopic dermatitis, particularly in children; psoriasis; pityriasis rubra pilaris; erythematous diseases triggered by various noxa, e.g., rays, chemicals, burns, etc.; bullous dermatoses; diseases of the lichenoid complex; pruritus (e.g., of allergic genesis); seborrheic dermatitis; rosacea; pemphigus vulgaris; erythema multiforme exudativum; balanitis; vulvitis; hair loss, such as occurs in alopecia areata; and cutaneous T cell lymphomas;
  • Dermatological diseases atopic dermatitis, particularly in children; psoriasis; pityriasis rubra pilaris; erythematous diseases triggered by various noxa, e.g., rays, chemicals, burns, etc.; bullous dermatoses
  • Renal diseases nephrotic syndrome; and all types of nephritis, e.g., glomerulonephritis;
  • Hepatic diseases acute liver cell disintegration; acute hepatitis of various genesis, e.g., viral, toxic, drug-induced; and chronically aggressive and/or chronically intermittent hepatitis;
  • Gastrointestinal diseases inflammatory bowel diseases, e.g., regional enteritis (Crohn disease), colitis ulcerosa; gastritis; peptic esophagitis (refluxoesophagitis); and gastroenteritis of other genesis, e.g., nontropical sprue;
  • Proctological diseases anal eczema; fissures; hemorrhoids; and idiopathic proctitis
  • Eye diseases allergic keratitis, uveitis, or LTDis; conjunctivitis; blepharitis; neuritis nervi optici; choroiditis; and sympathetic ophthalmia
  • Eye diseases allergic keratitis, uveitis, or ulceris; conjunctivitis; blepharitis; neuritis nervi optici; choroiditis; and sympathetic ophthalmia
  • Neurological diseases brain edema, particularly tumor-related brain edema; multiple sclerosis; acute encephalomyelitis; meningitis
  • Tumor diseases acute lymphatic leukemia; malignant lymphoma; lymphogranulomatoses; lymphosarcoma; extensive metastases, particularly in mammary, bronchial, and prostatic carcinoma;
  • various other disease-states or conditions including type I diabetes (insulin-dependent diabetes), osteoarthritis, Guillain-Barre syndrome, restenosis following percutaneous transluminal coronary angioplasty, Alzheimer disease, acute and chronic pain, atherosclerosis, reperfusion injury, bone resorption diseases, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, acute purulent meningitis, necrotizing enterocolitis and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion.
  • the compounds according to the invention can be used for the treatment of any other disease-states or conditions not mentioned above which have been treated, are treated, or will be treated with synthetic glucocorticoids (see, e.g., H.J. Hatz, Glucocorticoide: Immunologische Grundlagen, Pharmakologie und Therapierichtlinien [Glucocorticoids: Immunological Fundamentals, Pharmacology, and Therapeutic Guidelines], Stuttgart: Verlagsgesellschaft mbH, 1998, which is hereby incorporated by reference in its entirety). Most or all of the indications (i) through (xx) mentioned above are described in detail in H.J. Hatz, Glucocorticoide: Immunologische Grundlagen, Pharmakologie und Therapierichtlinien.
  • the compounds of the invention can also be used to treat disorders other than those listed above or mentioned or discussed herein, including in the Background of the Invention.
  • the antagonist compounds according to the invention can be used in patients as drugs for the treatment of the following disease-states or indications, without limitation: type II diabetes (non-insulin-dependent diabetes); obesity; cardiovascular diseases; hypertension; arteriosclerosis; neurological diseases, such as psychosis and depression; adrenal and pituitary tumors; glaucoma; and Cushing syndrome based on an ACTH-secreting tumor like pituitary adenoma.
  • type II diabetes non-insulin-dependent diabetes
  • obesity cardiovascular diseases
  • hypertension arteriosclerosis
  • neurological diseases such as psychosis and depression
  • adrenal and pituitary tumors glaucoma
  • Cushing syndrome based on an ACTH-secreting tumor like pituitary adenoma.
  • the compounds of the invention are useful for treating obesity and all disease-states and indications related to a deregulated fatty acids metabolism such as hypertension, atherosclerosis, and other cardiovascular diseases.
  • the antagonist compounds of the invention are useful in treating all disease-states and conditions that involve increased carbohydrate, protein, and lipid metabolism and would include disease-states and conditions leading to catabolism like muscle frailty (as an example of protein metabolism).
  • the compounds of the invention may also be used in diagnostic applications and for commercial and other purposes as standards in competitive binding assays.
  • the compounds of the invention may be used in the form of the compounds themselves or they may be modified by attaching a radioisotope, luminescence, fluorescent label or the like in order to obtain a radioisotope, luminescence, or fluorescent probe, as would be known by one of skill in the art and as outlined in Handbook of Fluorescent Probes and Research Chemicals, 6th Edition, R.P. Haugland (ed.), Eugene: Molecular Probes, 1996; Fluorescence and Luminescence Probes for Biological Activity, W.T. Mason (ed.), San Diego: Academic Press, 1993; Receptor-Ligand Interaction, A Practical Approach, E. C. Hulme (ed.), Oxford: IRL Press, 1992, each of which is hereby incorporated by reference in their entireties.
  • compositions When used as pharmaceuticals, the compounds of the invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared using procedures well known in the pharmaceutical art and comprise at least one compound of the invention.
  • the compounds of the invention may also be administered alone or in combination with adjuvants that enhance stability of the compounds of the invention, facilitate administration of pharmaceutical compositions containing them in certain embodiments, provide increased dissolution or dispersion, increased inhibitory activity, provide adjunct therapy, and the like.
  • the compounds according to the invention may be used on their own or in conjunction with other active substances according to the invention, optionally also in conjunction with other pharmacologically active substances.
  • the compounds of this invention are administered in a therapeutically or pharmaceutically effective amount, but may be administered in lower amounts for diagnostic or other purposes.
  • the compounds of the invention are useful in combination with glucocorticoids or corticosteroids.
  • standard therapy for a variety of immune and inflammatory disorders includes administration of corticosteroids, which have the ability to suppress immunologic and inflammatory responses.
  • corticosteroids While therapeutically beneficial, however, the use of corticosteroids is associated with a number of side effects, ranging from mild to possibly life threatening, especially with prolonged and/or high dose steroid usage. Accordingly, methods and compositions that enable the use of a lower effective dosage of corticosteroids (referred to as the "steroid sparing effect") would be highly desirable to avoid unwanted side effects.
  • the compounds of the invention provide such a steroid sparing effect by achieving the desired therapeutic effect while allowing the use of lower doses and less frequent administration of glucocorticoids or corticosteroids.
  • Administration of the compounds of the invention, in pure form or in an appropriate pharmaceutical composition can be carried out using any of the accepted modes of administration of pharmaceutical compositions.
  • administration can be, for example, orally, buccally (e.g., sublingually), nasally, parenterally, topically, transdermally, vaginally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as, for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
  • the pharmaceutical compositions will generally include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, adjuvants, diluents, vehicles, or combinations thereof.
  • Such pharmaceutically acceptable excipients, carriers, or additives as well as methods of making pharmaceutical compositions for various modes or administration are well-known to those of skill in the art. The state of the art is evidenced, e.g., by Remington: The Science and Practice of Pharmacy. 20th Edition, A. Gennaro (ed.), Lippincott Williams & Wilkins, 2000; Handbook of Pharmaceutical Additives, Michael & Irene Ash (eds.), Gower, 1995; Handbook of Pharmaceutical Excipients.
  • the forms of the compounds of the invention utilized in a particular pharmaceutical formulation will be selected (e.g., salts) that possess suitable physical characteristics (e.g., water solubility) that is required for the formulation to be efficacious.
  • compositions suitable for buccal (sub-lingual) administration include lozenges comprising a compound of the present invention in a flavored base, usually sucrose, and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
  • compositions suitable for parenteral administration comprise sterile aqueous preparations of a compound of the present invention. These preparations are preferably administered intravenously, although administration can also be effected by means of subcutaneous, intramuscular, or intradermal injection.
  • injectable pharmaceutical formulations are commonly based upon injectable sterile saline, phosphate-buffered saline, oleaginous suspensions, or other injectable carriers known in the art and are generally rendered sterile and isotonic with the blood.
  • the injectable pharmaceutical formulations may therefore be provided as a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, including 1,3-butanediol, water, Ringer's solution, isotonic sodium chloride solution, fixed oils such as synthetic mono- or diglycerides, fatty acids such as oleic acid, and the like.
  • a nontoxic parenterally acceptable diluent or solvent including 1,3-butanediol, water, Ringer's solution, isotonic sodium chloride solution, fixed oils such as synthetic mono- or diglycerides, fatty acids such as oleic acid, and the like.
  • injectable pharmaceutical formulations are formulated according to the known art using suitable dispersing or setting agents and suspending agents.
  • Injectable compositions will generally contain from 0.1 to 5% w/w of a compound of the invention.
  • Solid dosage forms for oral administration of the compounds include capsules, tablets, pills, powders, and granules.
  • a pharmaceutically acceptable composition containing a compound(s) of the invention is formed by the incorporation of any of the normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, pregelatinized starch, magnesium stearate, sodium saccharine, talcum, cellulose ether derivatives, glucose, gelatin, sucrose, citrate, propyl gallate, and the like.
  • Such solid pharmaceutical formulations may include formulations, as are well known in the art, to provide prolonged or sustained delivery of the drug to the gastrointestinal tract by any number of mechanisms, which include, but are not limited to, pH sensitive release from the dosage form based on the changing pH of the small intestine, slow erosion of a tablet or capsule, retention in the stomach based on the physical properties of the formulation, bioadhesion of the dosage form to the mucosal lining of the intestinal tract, or enzymatic release of the active drug from the dosage form.
  • Liquid dosage forms for oral administration of the compounds include emulsions, microemulsions, solutions, suspensions, syrups, and elixirs, optionally containing pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like. These compositions can also contain additional adjuvants such as wetting, emulsifying, suspending, sweetening, flavoring, and perfuming agents.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like.
  • additional adjuvants such as wetting, emulsifying, suspending, sweetening, flavoring, and perfuming agents.
  • Topical dosage forms of the compounds include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, eye ointments, eye or ear drops, impregnated dressings and aerosols, and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams. Topical application may be once or more than once per day depending upon the usual medical considerations.
  • preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles.
  • the formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation, more usually they will form up to about 80% of the formulation.
  • Transdermal administration is also possible.
  • Pharmaceutical compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • patches suitably contain a compound of the invention in an optionally buffered, aqueous solution, dissolved and/or dispersed in an adhesive, or dispersed in a polymer.
  • a suitable concentration of the active compound is about 1% to 35%, preferably about 3% to 15%.
  • the compounds of the invention are conveniently delivered in the form of an aerosol spray from a pump spray device not requiring a propellant gas or from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, heptafluoropropane, carbon dioxide, or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, heptafluoropropane, carbon dioxide, or other suitable gas.
  • the aerosol spray dosage unit may be determined by providing a valve to deliver a metered amount so that the resulting metered dose inhaler (MDI) is used to administer the compounds of the invention in
  • Such inhaler, nebulizer, or atomizer devices are known in the art, for example, in PCT International Publication Nos. WO 97/12687 (particularly Figure 6 thereof, which is the basis for the commercial RESPIMAT® nebulizer); WO 94/07607; WO 97/12683; and WO 97/20590, to which reference is hereby made and each of which is incorporated herein by reference in their entireties.
  • Rectal administration can be effected utilizing unit dose suppositories in which the compound is admixed with low-melting water-soluble or insoluble solids such as fats, cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights, or fatty acid esters of polyethylene glycols, or the like.
  • the active compound is usually a minor component, often from about 0.05 to 10% by weight, with the remainder being the base component.
  • the compounds of the invention are formulated with an acceptable carrier or excipient.
  • the carriers or excipients used must, of course, be acceptable in the sense of being compatible with the other ingredients of the composition and must not be deleterious to the patient.
  • the carrier or excipient can be a solid or a liquid, or both, and is preferably formulated with the compound of the invention as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compound.
  • Such carriers or excipients include inert fillers or diluents, binders, lubricants, disintegrating agents, solution retardants, resorption accelerators, absorption agents, and coloring agents.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or ⁇ -lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • a therapeutically effective daily dose is from about 0.001 mg to about 15 mg/kg of body weight per day of a compound of the invention; preferably, from about 0.1 mg to about
  • the dosage range would be from about 0.07 mg to about 1050 mg per day of a compound of the invention, preferably from about 7.0 mg to about 700 mg per day, and most preferably from about 7.0 mg to about 105 mg per day.
  • Some degree of routine dose optimization may be required to determine an optimal dosing level and pattern.
  • compositions encompass all the foregoing additives and the like.
  • the finely ground active substance, lactose, and some of the corn starch are mixed together.
  • the mixture is screened, then moistened with a solution of polyvinylpyrrolidone in water, kneaded, wet-granulated and dried.
  • the granules, the remaining corn starch and the magnesium stearate are screened and mixed together.
  • the mixture is compressed to produce tablets of suitable shape and size.
  • the finely ground active substance, some of the corn starch, lactose, microcrystalline cellulose, and polyvinylpyrrolidone are mixed together, the mixture is screened and worked with the remaining corn starch and water to form a granulate which is dried and screened.
  • the sodium-carboxymethyl starch and the magnesium stearate are added and mixed in and the mixture is compressed to form tablets of a suitable size.
  • the active substance, corn starch, lactose, and polyvinylpyrrolidone are thoroughly mixed and moistened with water.
  • the moist mass is pushed through a screen with a 1 mm mesh size, dried at about 45°C and the granules are then passed through the same screen.
  • convex tablet cores with a diameter of 6 mm are compressed in a tablet-making machine.
  • the tablet cores thus produced are coated in known manner with a covering consisting essentially of sugar and talc.
  • the finished coated tablets are polished with wax.
  • the substance and corn starch are mixed and moistened with water.
  • the moist mass is screened and dried.
  • the dry granules are screened and mixed with magnesium stearate.
  • the finished mixture is packed into size 1 hard gelatine capsules.
  • the active substance is dissolved in water at its own pH or optionally at pH 5.5 to 6.5 and sodium chloride is added to make it isotonic.
  • the solution obtained is filtered free from pyrogens and the filtrate is transferred under aseptic conditions into ampoules which are then sterilized and sealed by fusion.
  • the ampoules contain 5 mg, 25 mg, and 50 mg of active substance.
  • the hard fat is melted.
  • the ground active substance is homogeneously dispersed therein.
  • the mixture is cooled to 38°C and poured into slightly chilled suppository molds.
  • the suspension is transferred into a conventional aerosol container with a metering valve.
  • a metering valve Preferably, 50 ⁇ L of suspension are delivered per spray.
  • the active substance may also be metered in higher doses if desired (e.g., 0.02% by weight).

Abstract

Compounds of Formula (I), wherein R1, R2, R3, R4, R5, R6, X, R7, and R8 are as defined herein, or a tautomer, prodrug, solvate, or salt thereof; pharmaceutical compositions containing such compounds, and methods of modulating the glucocorticoid receptor function and methods of treating disease-states or conditions mediated by the glucocorticoid receptor function or characterized by inflammatory, allergic, or proliferative processes in a patient using these compounds.

Description

GLUCOCORTICOID MIMETICS, METHODS OF MAKING THEM, PHARMACEUTICAL COMPOSITIONS, AND USES THEREOF
Field of the Invention The present invention relates to glucocorticoid mimetics or ligands, methods of making such compounds, their use in pharmaceutical compositions, and their use in modulating the glucocorticoid receptor function, treating disease-states or conditions mediated by the glucocorticoid receptor function in a patient in need of such treatment, and other uses.
Background of the Invention
Glucocorticoids, a class of corticosteroids, are endogenous hormones with profound effects on the immune system and multiple organ systems. They suppress a variety of immune and inflammatory functions by inhibition of inflammatory cytokines such as IL-I, IL-2, IL-6, and TNF, inhibition of arachidonic acid metabolites including prostaglandins and leukotrienes, depletion of T-lymphocytes, and reduction of the expression of adhesion molecules on endothelial cells (PJ. Barnes, Clin. ScL, 1998, 94, pp. 557-572; P.J. Barnes et al, Trends Pharmacol. ScL, 1993, J_4, pp. 436-441). In addition to these effects, glucocorticoids stimulate glucose production in the liver and catabolism of proteins, play a role in electrolyte and water balance, reduce calcium absorption, and inhibit osteoblast function.
The anti-inflammatory and immune suppressive activities of endogenous glucocorticoids have stimulated the development of synthetic glucocorticoid derivatives including dexamethasone, prednisone, and prednisolone (L. Parente, Glucocorticoids. N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, pp. 35-54). These have found wide use in the treatment of inflammatory, immune, and allergic disorders including rheumatic diseases such as rheumatoid arthritis, juvenile arthritis, and ankylosing spondylitis, dermatological diseases including psoriasis and pemphigus, allergic disorders including allergic rhinitis, atopic dermatitis, and contact dermatitis, pulmonary conditions including asthma and chronic obstructive pulmonary disease (COPD), and other immune and inflammatory diseases including Crohn disease, ulcerative colitis, systemic lupus erythematosus, autoimmune chronic active hepatitis, osteoarthritis, tendonitis, and bursitis (J. Toogood, Glucocorticoids, N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, pp. 161-174). They have also been used to help prevent rejection in organ transplantation. Unfortunately, in addition to the desired therapeutic effects of glucocorticoids, their use is associated with a number of adverse side effects, some of which can be severe and life- threatening. These include alterations in fluid and electrolyte balance, edema, weight gain, hypertension, muscle weakness, development or aggravation of diabetes mellitus, and osteoporosis. Therefore, a compound that exhibited a reduced side effect profile while maintaining the potent anti-inflammatory effects would be particularly desirable, especially when treating a chronic disease.
The effects of glucocorticoids are mediated at the cellular level by the glucocorticoid receptor (R.H. Oakley and J. Cidlowski, Glucocorticoids, N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, pp. 55-80). The glucocorticoid receptor is a member of a class of structurally related intracellular receptors that when coupled with a ligand can function as a transcription factor that affects gene expression (R.M. Evans, Science, 1988, 240, pp. 889- 895). Other members of the family of steroid receptors include the mineralocorticoid, progesterone, estrogen, and androgen receptors. In addition to the effects mentioned above for glucocorticoids, hormones that act on this receptor family have a profound influence on body homeostasis, mineral metabolism, the stress response, and development of sexual characteristics. Glucocorticoids, N.J. Goulding and R.J. Flowers (eds.), Boston: Birkhauser, 2001, is hereby incorporated by reference in its entirety to better describe the state of the art.
A molecular mechanism which accounts for the beneficial anti-inflammatory effects and the undesired side effects has been proposed (e.g., S. Heck et al, EMBO J, 1994, J/7, pp. 4087- 4095; H.M. Reichardt et al, Cell, 1998, 93, pp. 531-541; F. Tranche et al, Curr. Opin. in Genetics and Dev., 1998, 8, pp. 532-538). Many of the metabolic and cardiovascular side effects are thought to be the result of a process called transactivation. In transactivation, the translocation of the ligand-bound glucocorticoid receptor to the nucleus is followed by binding to glucocorticoid response elements (GREs) in the promoter region of side effect- associated genes, for example, phosphoenolpyruvate carboxy kinase (PEPCK) in the case of increased glucose production. The result is an increased transcription rate of these genes which is believed to result, ultimately, in the observed side effects. The anti-inflammatory effects are thought to be due to a process called transrepression. In general, transrepression is a process independent of DNA binding that results from inhibition of NF -kB and AP-I- mediated pathways, leading to down regulation of many inflammatory and immune mediators. Additionally, it is believed that a number of the observed side effects may be due to the cross-reactivity of the currently available glucocorticoids with other steroid receptors, particularly the mineralocorticoid and progesterone receptors.
Thus, it may be possible to discover ligands for the glucocorticoid receptor that are highly selective and, upon binding, can dissociate the transactivation and transrepression pathways, providing therapeutic agents with a reduced side effect profile. Assay systems to determine effects on transactivation and transrepression have been described (e.g., CM. Bamberger and H.M. Schulte, Eur. J. Clin. Invest., 2000, 30 (suppl. 3), pp. 6-9). Selectivity for the glucocorticoid receptor may be determined by comparing the binding affinity for this receptor with that of other steroid family receptors including those mentioned above.
Glucocorticoids also stimulate the production of glucose in the liver by a process called gluconeogenesis and it is believed that this process is mediated by transactivation events. Increased glucose production can exacerbate type II diabetes, therefore a compound that selectivity inhibited glucocorticoid mediated glucose production may have therapeutic utility in this indication (J.E. Freidman et al, J. Biol. Chem., 1997, 272, pp. 31475-31481).
Novel ligands for the glucocorticoid receptor have been described in the scientific and patent literature. For example, PCT International Publication No. WO 99/33786 discloses triphenylpropanamide compounds with potential use in treating inflammatory diseases. PCT International Publication No. WO 00/66522 describes non-steroidal compounds as selective modulators of the glucocorticoid receptor potentially useful in treating metabolic and inflammatory diseases. PCT International Publication No. WO 99/41256 describes tetracyclic modulators of the glucocorticoid receptor potentially useful in treating immune, autoimmune, and inflammatory diseases. U.S. Patent No. 5,688,810 describes various non-steroidal compounds as modulators of glucocorticoid and other steroid receptors. PCT International Publication No. WO 99/63976 describes a non-steroidal, liver-selective glucocorticoid antagonist potentially useful in the treatment of diabetes. PCT International Publication No. WO 00/32584 discloses non-steroidal compounds having anti-inflammatory activity with dissociation between anti-inflammatory and metabolic effects. PCT International Publication No. WO 98/54159 describes non-steroidal cyclically substituted acylanilides with mixed gestagen and androgen activity. U.S. Patent No. 4,880,839 describes acylanilides having progestational activity and EP 253503 discloses acylanilides with antiandrogenic properties. PCT International Publication No. WO 97/27852 describes amides that are inhibitors of farnesylprotein transferase.
A compound that is found to interact with the glucocorticoid receptor in a binding assay could be an agonist or an antagonist. The agonist properties of the compound could be evaluated in the transactivation or transrepression assays described above. Given the efficacy demonstrated by available glucocorticoid drugs in inflammatory and immune diseases and their adverse side effects, there remains a need for novel glucocorticoid receptor agonists with selectivity over other members of the steroid receptor family and a dissociation of the transactivation and transrepression activities. Alternatively, the compound may be found to have antagonist activity. As mentioned above, glucocorticoids stimulate glucose production in the liver. Increased glucose production induced by glucocorticoid excess can exacerbate existing diabetes, or trigger latent diabetes. Thus a ligand for the glucocorticoid receptor that is found to be an antagonist may be useful, inter alia, for treating or preventing diabetes.
Summary of the Invention
The instant invention is directed to compounds of Formula (IA)
Figure imgf000005_0001
wherein:
R1 is hydrogen or Ci-C3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo;
R2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups, wherein each substituent group of R2 is independently C1-Cs alkyl, C2-C3 alkenyl, C2- C3 alkynyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C1-Cs alkoxycarbonylamino, C1-Cs alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-
C5 alkyl; or ureido wherein either nitrogen atom is optionally independently substituted with C1-C5 alkyl; or C1-C5 alkylthio,
wherein each substituent group of R2 is optionally independently substituted with C1-C3 alkyl, halogen, hydroxyl, or amino,
wherein R2 cannot be /?-methylphenyl;
R3 is a hydrogen or C1-C5 alkyl, optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R3 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R4 and R5 are each independently hydrogen, C1-C5 alkyl, or phenyl, or R4 and R5 together with the carbon atom they are commonly attached to form a C3-C8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R4 and R5 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R6 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R6 is independently C1-C3 alkyl, C2-C5 alkenyl, C2-
C5 alkynyl, heterocyclyl, aryl, heteroaryl, C1-C5 alkoxy, acyl, acylamino, aminocarbonyl, C1-C3 alkylamioncarbonyl, C1-C3 dialkylaminocarbonyl halogen, hydroxy, carboxy, cyano, oxo, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, nitro, or amino wherein the nitrogen atom is optionally independently mono- or di- substituted by C1-Cs alkyl; or C1-Cs alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R6 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, halogen, hydroxy, oxo, cyano, phenyl, amino, or trifluoromethyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, phenyl optionally substituted with Ci-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
One aspect of the invention includes compounds of Formula (IA), wherein:
R1 is hydrogen;
R2 is phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently Ci-C3 alkyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-Cs alkyl, or Ci-C5 alkylthio, wherein R2 cannot be p-methylphenyl;
R3 is hydrogen;
R4 and R5 are each hydrogen or C1-Cs alkyl;
R6 is an indolyl, dihydroisoindolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, dihydroisobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, dihydrobenzodioxepinyl, acridinyl, pyrimidinyl or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R6 is independently Ci-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C1-C3 alkoxy, acylamino, aminocarbonyl, C1-C3 alkylaminocarbonyl, C1-C3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or Ci-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, phenyl, or trifluoromethyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, phenyl, hydroxy, oxo, cyano, amino, or trifluoromethyl, or a tautomer, prodrug, solvate, or salt thereof.
Another aspect of the invention includes compounds of Formula (IA), wherein:
R1 is hydrogen;
R2 is a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-Cs alkyl, C1-C3 alkoxy, hydroxy, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-C3 alkyl, or Ci-C3 alkylthio,
wherein R2 cannot be />-methylphenyl;
R3 is hydrogen;
R4 and R5 are each hydrogen;
R6 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R6 is independently Ci-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, Ci-C3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or Ci-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, wherein each substituent group of R6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl;
5 X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, C1-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one,
10 two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, phenyl, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
15 The following are representative compounds of Formula (I) according to the invention:
Compound Name Compound Structure
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-5- yloxymethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-8- yloxymethyl)ethyl]benzenesulfonamide
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-4- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(pyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3- trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-5- ylaminomethyl)ethyl]benzenesulfonamide
Figure imgf000015_0001
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-6- ylaminomethyl)ethyl]benzenesulfonamide
N-{l-[(2,3-Dihydro-l,4-benzodioxin-6- ylamino)methyl]-2,2,2-trifluoroethyl}-2,4,6- trimethylbenzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(isoquinolin- 5-ylaminomethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-{2,2,2-trifluoro-l-[(2- methy lquinolin- 8 - ylamino)methyl]ethyl}benzenesulfonamide
N-[I -(Benzothiazol-7-ylaminomethyl)-2,2,2- trifluoroethyl]-2,4,6-trimethylbenzenesulfonamide
Figure imgf000016_0001
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(2-methyl- l,3-dioxo-2,3-dihydro-lH-isoindol-5- ylamino)methyl]ethyl}benzenesulfonamide
N-{l-[(l,l-Dioxo-lH-lλ6-benzo[ό]thiophen-5- ylamino)methyl]-2,2,2-trifluoroethyl}-2,4,6- trimethylbenzenesulfonamide
N- { 1 -[(3,4-Dihydro-2H- 1 ,5-benzodioxepin-7- ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -
[(methylpyridin-2- ylamino)methyl]ethyl}benzenesulfonamide
2,4,6-Trimethyl-N-(2,2,2-trifluoro-l-{[(4- methoxybenzyl)pyridin-2- ylamino]methyl}ethyl)benzenesulfonamide
Figure imgf000017_0001
N-( 1 - { [Benzyl-(4-methylpyridin-2-yl)- amino]methyl}-2,2,2-trifluoroethyl)-2,4,6- trimethylbenzenesulfonamide
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -
[(methylpyridin-4- ylamino)methyl]ethyl}benzenesulfonamide
2,4,6-Trimetriyl-N-(2,2,2-trifluoro-l-{[(6- methoxypyridin-2- yl)methylamino]methyl}ethyl)benzenesulfonamide
N-[I -(4,6-Dimethylpyrimidin-2-ylsulfanylmethyl)-
2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(2-oxo-2,3- dihydro- lH-benzimidazol-5- ylamino)methyl]ethyl}benzenesulfonamide
Figure imgf000018_0001
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-thiophen-
2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(quinolin-4- yloxymethyl)ethyl]benzenesulfonamide
Figure imgf000019_0001
or a tautomer, prodrug, solvate, or salt thereof.
Preferred compounds of Formula (IA) include the following:
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-8-yloxymethyl)ethyl]benzenesulfonamide; 5
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-7-yloxymethyl)ethyl]benzenesulfonamide; 0 N-[I -(Acridin-4-yloxymethyl)-2,2,2-trifluoroethyl]-2,4,6-trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-oxo-2,3-dihydrobenzofuran-6- yloxymethyl)ethyl]benzenesulfonamide;
5 2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(isoquinolin-7-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-8-ylsulfanylmethyl)ethyl]benzenesulfonamide; 2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-methylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-methyl-2-oxo-2H- 1 -benzopyran-7- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-furan-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
N-[l-(3-Cyano-6-methyl-4-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]- 2,4,6-trimethylbenzenesulfonamide;
N-[l-(3-Chloro-5-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(5-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-trifluoromethylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[4-(4-methoxyphenyl)pyrimidin-2- ylsulfanylmethyl] ethyl } benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-phenoxypyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(5-nitropyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide; 2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-4-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(pyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-ylaminomethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-6-ylaminomethyl)ethyl]benzenesulfonamide;
N- { 1 -[(2,3-Dihydro- 1 ,4-benzodioxin-6-ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-ylaminomethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(2-methylquinolin-8- ylamino)methyl]ethyl}benzenesulfonamide;
N-[l-(Benzothiazol-7-ylaminomethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide;
N- { 1 -[(1 , 1 -Dioxo- IH- lλ6-benzo[Z?]thiophen-5-ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide;
N-{l-[(3,4-Dihydro-2H-l,5-benzodioxepin-7-ylamino)methyl]-2,2,2-trifluoroethyl}-2,4,6- trimethylbenzenesulfonamide; N-[l-(4,6-Dimethylpyrimidin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide ;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(4-thiophen-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide; and
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-4-yloxymethyl)ethyl]benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
More preferred compounds of Formula (IA) include the following:
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(isoquinolin-5-yloxymethyl)ethyl]benzenesulfonamide ;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-oxo-2,3-dihydrobenzofuran-6- yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-7-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-8-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-methyl-2-oxo-2H- 1 -benzopyran-7- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-furan-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
N-[l-(3-Cyano-6-methyl-4-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]- 2,4,6-trimethylbenzenesulfonamide;
N-[l-(3-Chloro-5-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide; 2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(5-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-trifluoromethylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-phenoxypyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(5-nitropyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-ylaminomethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-6-ylaminomethyl)ethyl]benzenesulfonamide;
N- {l-[(2,3-Dihydro-l,4-benzodioxin-6-ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-ylaminomethyl)ethyl]- benzenesulfonamide;
2,4,6-Trimethyl-N-{2,2,2-trifluoro-l-[(2-methylquinolin-8-ylamino)methyl]ethyl}- benzenesulfonamide; N-[l-(Benzothiazol-7-ylaminomethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide ;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-thiophen-2-ylpyrimidin-2-ylsulfanylmethyl)ethyl]- benzenesulfonamide; and
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-4-yloxymethyl)ethyl]benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
Most preferred compounds of Formula (IA) include the following:
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
N-[l-(3-Cyano-6-methyl-4-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]- 2,4,6-trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(5-nitropyridin-2-ylsulfanylmethyl)ethyl]- benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-trifluoromethylpyridin-2-ylsulfanylmethyl)ethyl]- benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-ylaminomethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-ylaminomethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N-{2,2,2-trifluoro-l-[(2-methylquinolin-8-ylamino)methyl]ethyl}- benzenesulfonamide; and 2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-4-yloxymethyl)ethyl]benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
The invention also provides a method of making a compound of Formula (IA)
Figure imgf000025_0001
where R1 is H and R2, R3, R4, R5, R6, and X are as defined above, the method comprising reacting an aziridine compound of Formula (II) with a reagent R6X-M of Formula (III) where M is Na, K, or Li, or where X is nitrogen or sulfur and M is hydrogen, in a suitable solvent to form the compound of Formula (IA)
Figure imgf000025_0002
IA
The invention further provides a method of making a compound of Formula (IA)
Figure imgf000025_0003
where R1, R3, R4, and R5 are each H, and R2 and R6 are as defined above, the method comprising:
(a) reacting the amino acid ester where R' is methyl or ethyl of Formula (IV) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as dichloromethane, in the presence of a base, such as triethylamine, or in pyridine to form an sulfonamide of
Formula (VI)
Figure imgf000026_0001
IV V Vl
(b) reacting the carboxylic acid ester of Formula (VI) with a reducing agent, such as lithium aluminum hydride, in a suitable solvent, such as ether or tetrahydrofuran, to form an alcohol of Formula (VII)
Figure imgf000026_0002
Vl VII
(c) reacting the alcohol of Formula (VII) with a sulfonyl chloride, such as methane sulfonyl chloride or /?-toluenesulfonyl chloride, in a suitable solvent, such as tetrahydrofuran, in the presence of a suitable base, such as sodium hydride, to form an aziridine of Formula (II) where R3, R4, and R5 are each H
Figure imgf000026_0003
VII Il .
(d) reacting an aziridine of Formula (II) with a reagent of R6X-M of Formula (III) where X is sulfur, oxygen or NR7 and M is Na, K, or Li, or where X is NR7 or sulfur and M is hydrogen, in a suitable solvent to form the compound of Formula (IA)
Figure imgf000026_0004
Il III IA
A second method for making a compound of Formula (IA) comprises:
(a') reacting an amine of Formula (VIII) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as pyridine, to form an sulfonamide of Formula (IX)
Figure imgf000027_0001
VIII V IX
(b') reacting a thiol of Formula (IX) with a oxonium salt, such as trimethyloxonium tetrafluoroborate, in a suitable solvent, such as dichloromethane, to form a sulfonium salt of Formula (X)
Figure imgf000027_0002
IX X
(c') cyclizing the sulfonium salt of Formula (X) in a suitable solvent, such as tetrahydrofuran, in the presence of a suitable base, such as sodium hydride, to form an aziridine of Formula (II) where R3, R4, and R5 are each H which may be converted to a compound of Formula (IA) as shown above
Cyclization
Figure imgf000027_0003
Figure imgf000027_0004
Alternatively, the group R2 may be substituted with another R2 group, the method comprising:
(a") reacting a sulfonamide when R2 is an ørt/zø-substituted nitrophenyl with a thiol such as thiophenol in the presence of a base such as potassium carbonate in DMF to form an amine of Formula (XI)
Figure imgf000027_0005
IA Xl and (b") reacting the aminoethyl compound of Formula (XI) with a sulfonyl halide of Formula (V) in the presence of a suitable base and solvent, such as pyridine, to form the compound of Formula (IA) where R2 is not ørt/zø-nitrophenyl
B DadsbetJ
Figure imgf000028_0001
Figure imgf000028_0002
Xl IA
The invention is also directed to compounds of Formula (IB)
Figure imgf000028_0003
wherein:
R1 is hydrogen or Ci-C3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo;
R2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently Ci-C5 alkyl, C2-C3 alkenyl, C2- C3 alkynyl, C1-C5 alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C1-C5 alkoxycarbonylamino, C1-C5 alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1- C5 alkyl; or ureido wherein either nitrogen atom is optionally independently substituted with C1-C5 alkyl; or C1-C5 alkylthio,
wherein each substituent group of R is optionally independently substituted with Ci- C3 alkyl, halogen, hydroxyl, or amino, wherein R2 cannot be p-methylphenyl;
R3 is Ci-Cs alkyl independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R3 is independently C3-C6 cycloalkyl, aryl, halogen, trifluoromethyl, trifluoromethoxy, or trifluoromethylthio,
wherein R cannot be a trifluoromethyl;
R4 is a hydrogen or C1-Cs alkyl, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R4 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
wherein R4 cannot be a trifluoromethyl;
R5 and R6 are each independently hydrogen, C1-Cs alkyl or phenyl or R5 and R6 together with the carbon atom they are commonly attached to form a C3-C8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R5 and R6 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and R8 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently C1-C3 alkyl, C2-C5 alkenyl, C2-
C5 alkynyl, heterocyclyl, aryl, heteroaryl, C1-Cs alkoxy, acyl, acylamino, aminocarbonyl, C1-C3 alkylamioncarbonyl, C1-C3 dialkylaminocarbonyl halogen, hydroxy, carboxy, cyano, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, nitro, or amino wherein the nitrogen atom is optionally independently mono- or di- substituted by C1-Cs alkyl; or C1-Cs alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R8 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
One aspect of the invention includes compounds of Formula (IB), wherein:
R1 is hydrogen;
R2 is phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-C3 alkyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-Cs alkyl, or Ci-C5 alkylthio,
wherein R2 cannot be /?-methylphenyl; R3 is Ci-C5 alkyl independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R3 is independently C3-C8 cycloalkyl, halogen, trifluoromethyl, or trifluoromethoxy,
wherein R3 cannot be a trifluoromethyl;
R4 is hydrogen;
R5 and R6 are each hydrogen or C1-Cs alkyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, C1-C5 alkyl, or phenyl;
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and
R8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently Ci-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, Ci-C3 alkoxy, acylamino, aminocarbonyl, Ci-C3 alkylaminocarbonyl, Ci-C3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, trifluoromethyl, or Ci-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, wherein each substituent group of R8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl ,
or a tautomer, prodrug, solvate, or salt thereof.
Another aspect of the invention includes compounds of Formula (IB), wherein:
R1 is hydrogen;
R2 is a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R ,2 i •s independently C1-Cs alkyl, C1-C3 alkoxy, hydroxy, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-C3 alkyl, or Ci-C3 alkylthio;
wherein R2 can not be /?-methylphenyl;
R3 is methyl, ethyl, isopropyl, or tert-butyl;
R4 is hydrogen;
R5 and R6 are each hydrogen;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, Ci-C5 alkyl, or phenyl, wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently C1-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C1-C3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, trifluoromethyl, or C1-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
The following are representative compounds of Formula (IB) according to the invention:
Compound Name Compound Structure
N-{(5)-l-[(lH-Indol-7- ylamino)methyl]propyl} -2,4,6- trimethylbenzenesulfonamide
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Preferred compounds of Formula (IB) include the following:
N-{(5)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-4-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide N-{(5)-l-[(lH-Indol-6-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-5-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-((5)-l-{[l-(4-Fluorophenyl)-lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide ;
N-{(5)-l-[(lH-Indazol-5-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N- {(5)- 1 -[( lH-Indazol-6-ylamino)methyl]propyl} -2,4,6-trimethylbenzenesulfonamide;
2-Amino-4,6-dichloro-N-((S)- 1 - { [ 1 -(4-fluorophenyl)- lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide; and
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
More preferred compounds of Formula (IB) include the following:
N-{(5)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-4-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-6-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-5-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-((5)-l-{[l-(4-Fluorophenyl)-lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indazol-6-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide; 2-Amino-4,6-dichloro-N-((5)- 1 - { [ 1 -(4-fluorophenyl)- lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide; and
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
Most preferred compounds of Formula (IB) include the following:
N-{(5)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-4-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-(OS)- 1 - { [ 1 -(4-Fluorophenyl)- lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide;
2-Amino-4,6-dichloro-N-((5)- 1 - { [ 1 -(4-fluorophenyl)- lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide; and
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
The invention also provides a method of making a compound of Formula (IB)
Figure imgf000037_0001
wherein R1, R4, R5, and R6 are each Η, and R2, R3, R8, and X are as defined above, the method comprising: (a) reacting the amino alcohol of Formula (XII) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as tetrahydrofuran, in the presence of a base, such as sodium hydride, or in dichloromethane in the presence of pyridine followed by a suitable base, such as aqueous potassium hydroxide, to form an aziridine of Formula (XIII)
Figure imgf000038_0001
x" v xι" ; and
(b) reacting the aziridine of Formula (XIII) with an organometallic reagent R8X-M of Formula (III) where M is Na, Li, or MgX and X is Cl, Br, or I, in a suitable solvent such as ether, tetrahydrofuran, DMF, or ethanol, to form the compound of Formula (IB)
Figure imgf000038_0002
XIII III IB . or (c) reacting the aziridine of Formula (XIII) with an aniline or thiol (X is NR7 or S) reagent R8-XH of Formula (XIV) in a suitable solvent, such as tetrahydrofuran, methanol, or acetonitrile, under thermal conditions with or without an additive such as lithium perchlorate, β-cyclodextrin hydrate, or triethylamine to form the compound of Formula (IB)
Figure imgf000038_0003
XIII XIV IB
Alternatively, the group R2 may be substituted with another R2 group, the method comprising:
(a') reacting a sulfonamide where R2 is an ortho nitrophenyl group of formula (IB) with a thiol such as thiophenol in the presence of a base, such as potassium carbonate in DMF, to form an amino compound of Formula (XV)
Figure imgf000039_0001
IB XV ; and
(b') reacting the aminoethyl compound of Formula (XV) with a sulfonyl halide of Formula (V) in the presence of a suitable base, such as triethylamine or pyridine, in a suitable solvent, such as dichloromethane, to form the compound of Formula (IB) where R is not an ortho nitrophenyl group
Figure imgf000039_0002
XV V IB
The intermediate aziridine of Formula (XIII) may be made by the following method:
(a) reacting the amino acid ester where R' is methyl or ethyl of Formula (XVI) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as dichloromethane, in the presence of a base, such as triethylamine, or in pyridine to form an sulfonamide of Formula (XVII)
Figure imgf000039_0003
XVI V XVII
(b) reacting the carboxylic acid ester of Formula (XVI) with a reducing agent such as lithium aluminum hydride in a suitable solvent, such as ether or tetrahydrofuran, to form an alcohol of Formula (XVIII)
Figure imgf000039_0004
XVII XVIII ; and (c) reacting the alcohol of Formula (XVIII) with a sulfonyl chloride, such as methane sulfonyl chloride or p-toluenesulfonyl chloride, in a suitable solvent, such as tetrahydrofuran, in the presence of a suitable base, such as sodium hydride, to form an aziridine of Formula (XIII)
H
I Cyclization
HO' ^'^S-
FT *FT
R° O R° O XVIII XIII
Alternatively, the intermediate aziridine of Formula (XIII) may be made by the following method:
(a") reacting the amino alcohol of Formula (XII) with a sulfonyl chloride of Formula (V) in a suitable solvent, such as dichloromethane, in the presence of a base, such as triethylamine or pyridine, to form a compound of Formula (XVIII)
^<=>2
Figure imgf000040_0001
XII V XVIII ; and
(b") reacting the alcohol of Formula (XVIII) with a sulfonyl chloride, such as methane sulfonyl chloride or /?βrø-toluenesulfonyl chloride, in a suitable solvent, such as tetrahydrofuran, in the presence of a suitable base, such as sodium hydride, to form an aziridine of Formula (XIII)
H
I Cyclization
Ho I 3 N-s I lt° R2 "r I 3 N^ Ilt° R
R O R O
XVIII XIII
In another aspect of the invention, the compounds according to the invention are formulated into pharmaceutical compositions comprising an effective amount, preferably a pharmaceutically effective amount, of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof, and a pharmaceutically acceptable excipient or carrier. The invention also provides a method of modulating the glucocorticoid receptor function in a patient, the method comprising administering to the patient an effective amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
The invention further provides a method of treating a disease-state or condition mediated by the glucocorticoid receptor function in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
In addition, the invention also provides a method of treating a disease-state or condition selected from: type II diabetes, obesity, cardiovascular diseases, hypertension, arteriosclerosis, neurological diseases, adrenal and pituitary tumors, and glaucoma, in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof.
The invention provides a method of treating a disease characterized by inflammatory, allergic, or proliferative processes, in a patient in need of such treatment, the method comprising administering to the patient an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof. In a preferred embodiment of the invention, the disease characterized by inflammatory, allergic, or proliferative processes is selected from: (i) lung diseases; (ii) rheumatic diseases or autoimmune diseases or joint diseases; (iii) allergic diseases; (iv) vasculitis diseases; (v) dermatological diseases; (vi) renal diseases; (vii) hepatic diseases; (viii) gastrointestinal diseases; (ix) proctological diseases; (x) eye diseases; (xi) diseases of the ear, nose, and throat (ENT) area; (xii) neurological diseases; (xiii) blood diseases; (xiv) tumor diseases; (xv) endocrine diseases; (xvi) organ and tissue transplantations and graft-versus-host diseases; (xvii) severe states of shock; (xviii) substitution therapy; and (xix) pain of inflammatory genesis. In another preferred embodiment of the invention, the disease characterized by inflammatory, allergic, or proliferative processes is selected from: type I diabetes, osteoarthritis, Guillain-Barre syndrome, restenosis following percutaneous transluminal coronary angioplasty, Alzheimer disease, acute and chronic pain, atherosclerosis, reperfusion injury, bone resorption diseases, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, acute purulent meningitis, necrotizing enterocolitis, and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion.
The invention further provides methods of treating the disease-states or conditions mentioned above, in a patient in need of such treatment, the methods comprising sequentially or simultaneously administering to the patient: (a) an effective amount of a pharmaceutically acceptable compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) a pharmaceutically acceptable glucocorticoid.
The invention further provides a method of assaying the glucocorticoid receptor function in a sample, comprising: (a) contacting the sample with a selected amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) detecting the amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof bound to glucocorticoid receptors in the sample. In a preferred embodiment of the invention, the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof is labeled with a detectable marker selected from: a radiolabel, fluorescent tag, a chemiluminescent tag, a chromophore, and a spin label.
The invention also provides a method of imaging the glucocorticoid receptor distribution in a sample or patient, the method comprising: (a) contacting the sample or administering to a patient a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker; (b) detecting the spatial distribution and amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker bound to glucocorticoid receptors in the sample or patient using an imaging means to obtain an image; and (c) displaying an image of the spatial distribution and amount of the compound according to the invention or a tautomer, prodrug, solvate, or salt thereof having a detectable marker bound to glucocorticoid receptors in the sample. In a preferred embodiment of the invention, the imaging means is selected from: radioscintigraphy, nuclear magnetic resonance imaging (MRI), computed tomography (CT scan), or positron emission tomography (PET). The invention also provides a kit for the in vitro diagnostic determination of the glucocorticoid receptor function in a sample, comprising: (a) a diagnostically effective amount of a compound according to the invention or a tautomer, prodrug, solvate, or salt thereof; and (b) instructions for use of the diagnostic kit.
Definition of Terms and Conventions Used
Terms not specifically defined herein should be given the meanings that would be given to them by one of skill in the art in light of the disclosure and the context. As used in the specification and appended claims, however, unless specified to the contrary, the following terms have the meaning indicated and the following conventions are adhered to.
A. Chemical Nomenclature, Terms, and Conventions
In the groups, radicals, or moieties defined below, the number of carbon atoms is often specified preceding the group, for example, Ci-Ci0 alkyl means an alkyl group or radical having 1 to 10 carbon atoms. The term "lower" applied to any carbon-containing group means a group containing from 1 to 8 carbon atoms, as appropriate to the group (i.e., a cyclic group must have at least 3 atoms to constitute a ring). In general, for groups comprising two or more subgroups, the last named group is the radical attachment point, for example, "alkylaryl" means a monovalent radical of the formula AIk-Ar-, while "arylalkyl" means a monovalent radical of the formula Ar-AIk- (where AIk is an alkyl group and Ar is an aryl group). Furthermore, the use of a term designating a monovalent radical where a divalent radical is appropriate shall be construed to designate the respective divalent radical and vice versa. Unless otherwise specified, conventional definitions of terms control and conventional stable atom valences are presumed and achieved in all formulas and groups.
The terms "alkyl" or "alkyl group" mean a branched or straight-chain saturated aliphatic hydrocarbon monovalent radical. This term is exemplified by groups such as methyl, ethyl, n-propyl, 1-methylethyl (isopropyl), n-butyl, n-pentyl, 1,1 -dimethyl ethyl (tert-butyl), and the like. It may be abbreviated "AIk". The terms "alkenyl" or "alkenyl group" mean a branched or straight-chain aliphatic hydrocarbon monovalent radical containing at least one carbon-carbon double bond. This term is exemplified by groups such as ethenyl, propenyl, n-butenyl, isobutenyl, 3-methylbut- 2-enyl, n-pentenyl, heptenyl, octenyl, decenyl, and the like.
The terms "alkynyl" or "alkynyl group" mean a branched or straight-chain aliphatic hydrocarbon monovalent radical containing at least one carbon-carbon triple bond. This term is exemplified by groups such as ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, n- pentynyl, heptynyl, octynyl, decynyl, and the like.
The terms "alkylene" or "alkylene group" mean a branched or straight-chain saturated aliphatic hydrocarbon divalent radical having the specified number of carbon atoms. This term is exemplified by groups such as methylene, ethylene, propylene, n-butylene, and the like, and may alternatively and equivalently be denoted herein as -(alkyl)-.
The terms "alkenylene" or "alkenylene group" mean a branched or straight-chain aliphatic hydrocarbon divalent radical having the specified number of carbon atoms and at least one carbon-carbon double bond. This term is exemplified by groups such as ethenylene, propenylene, n-butenylene, and the like, and may alternatively and equivalently be denoted herein as -(alkylenyl)-.
The terms "alkynylene" or "alkynylene group" mean a branched or straight-chain aliphatic hydrocarbon divalent radical containing at least one carbon-carbon triple bond. This term is exemplified by groups such as ethynylene, propynylene, n-butynylene, 2-butynylene, 3- methylbutynylene, n-pentynylene, heptynylene, octynylene, decynylene, and the like, and may alternatively and equivalently be denoted herein as -(alkynyl)-.
The terms "alkoxy" or "alkoxy group" mean a monovalent radical of the formula AIkO-, where AIk is an alkyl group. This term is exemplified by groups such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, sec-butoxy, tert-butoxy, pentoxy, and the like. The terms "aryloxy", "aryloxy group", mean a monovalent radical of the formula ArO-, where Ar is aryl. This term is exemplified by groups such as phenoxy, naphthoxy, and the like.
The terms "alkylcarbonyl", "alkylcarbonyl group", "alkanoyl", or "alkanoyl group" mean a monovalent radical of the formula AIkC(O)-, where AIk is alkyl or hydrogen.
The terms "arylcarbonyl", "arylcarbonyl group", "aroyl" or "aroyl group" mean a monovalent radical of the formula ArC(O)-, where Ar is aryl.
The terms "acyl" or "acyl group" mean a monovalent radical of the formula RC(O)-, where R is a substituent selected from hydrogen or an organic substituent. Exemplary substituents include alkyl, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, and the like. As such, the terms comprise alkylcarbonyl groups and arylcarbonyl groups.
The terms "acylamino" or "acylamino group" mean a monovalent radical of the formula RC(O)N(R)-, where each R is a substituent selected from hydrogen or a substituent group.
The terms "alkoxycarbonyl" or "alkoxycarbonyl group" mean a monovalent radical of the formula AIkO-C(O)-, where AIk is alkyl. Exemplary alkoxycarbonyl groups include methoxycarbonyl, ethoxycarbonyl, tert-butyloxycarbonyl, and the like.
The terms "aryloxycarbonyl" or "aryloxycarbonyl group" mean a monovalent radical of the formula ArO-C(O)-, where Ar is aryl.
The terms "alkylcarbonyloxy" or "alkylcarbonyloxy group" or "alkanoyloxy" or "alkanoyloxy group" mean a monovalent radical of the formula AIkC(O)O-, where AIk is alkyl.
The terms "arylcarbonyloxy" or "arylcarbonyloxy group" or "aroyloxy" or "aroyloxy group" mean a monovalent radical of the formula ArC(O)O-, where Ar is aryl. The terms "alkylaminocarbonyloxy" or "alkylaminocarbonyloxy group" mean a monovalent radical of the formula R2NC(O)O-, where each R is independently hydrogen or lower alkyl.
The term "alkoxycarbonylamino" or "alkoxycarbonylamino group" mean a monovalent radical of the formula ROC(O)NH-, where R is lower alkyl.
The terms "alkylcarbonylamino" or "alkylcarbonylamino group" or "alkanoylamino" or "alkanoylamino groups" mean a monovalent radical of the formula AIkC(O)NH-, where AIk is alkyl. Exemplary alkylcarbonylamino groups include acetamido (CHsC(O)NH-).
The terms "alkylaminocarbonyloxy" or "alkylaminocarbonyloxy group" mean a monovalent radical of the formula AIkNHC(O)O-, where AIk is alkyl.
The terms "amino" or "amino group" mean an -NH2 group.
The terms "alkylamino" or "alkylamino group" mean a monovalent radical of the formula (AIk)NH-, where AIk is alkyl. Exemplary alkylamino groups include methylamino, ethylamino, propylamino, butylamino, tert-butylamino, and the like.
The terms "dialkylamino" or "dialkylamino group" mean a monovalent radical of the formula (AIk)(AIk)N-, where each AIk is independently alkyl. Exemplary dialkylamino groups include dimethylamino, methylethylamino, diethylamino, dipropylamino, ethylpropylamino, and the like.
The terms "substituted amino" or "substituted amino group" mean a monovalent radical of the formula -NR2, where each R is independently a substituent selected from hydrogen or the specified substituents (but where both Rs cannot be hydrogen). Exemplary substituents include alkyl, alkanoyl, aryl, arylalkyl, cycloalkyl, heterocyclyl, heteroaryl, heteroarylalkyl, and the like.
The terms "alkoxycarbonylamino" or "alkoxycarbonylamino group" mean a monovalent radical of the formula AIkOC(O)NH-, where AIk is alkyl. The terms "ureido" or "ureido group" mean a monovalent radical of the formula R.2NC(O)NH-, where each R is independently hydrogen or alkyl.
The terms "halogen" or "halogen group" mean a fluoro, chloro, bromo, or iodo group.
The term "halo" means one or more hydrogen atoms of the group are replaced by halogen groups.
The terms "haloalkyl" or "haloalkyl group" mean a branched or straight-chain saturated aliphatic hydrocarbon monovalent radical, wherein one or more hydrogen atoms thereof are each independently replaced with halogen atoms. This term is exemplified by groups such as chloromethyl, 1 ,2-dibromoethyl, 1 , 1 , 1 -trifluoropropyl, 2-iodobutyl, l-chloro-2-bromo-3- fluoropentyl, and the like.
The terms "sulfanyl", "sulfanyl group", "thioether", or "thioether group" mean a divalent radical of the formula -S-.
The terms "alkylthio" or "alkylthio group" mean a monovalent radical of the formula AIkS-, where AIk is alkyl. Exemplary groups include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, and the like.
The terms "arylthio" or "arylthio group" mean a monovalent radical of the formula ArS-, where Ar is aryl.
The terms "sulfinyl", "sulfinyl group", "thionyl", or "thionyl group" mean a divalent radical of the formula -SO-.
The terms "sulfonyl" or "sulfonyl group" mean a divalent radical of the formula -SO2-.
The terms "sulfonylamino" or "sulfonylamino group" mean a divalent radical of the formula -SO2NR-, where R is a hydrogen or a substituent group. The terms "aminosulfonyl" or "aminosulfonyl group" mean a monovalent radical of the formula NR2SO2-, where R is each independently a hydrogen or a substituent group.
The terms "carbocycle" or "carbocyclic group" mean a stable aliphatic 3- to 15-membered monocyclic or polycyclic monovalent or divalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the carbocycle may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. The term comprises cycloalkyl (including spiro cycloalkyl), cycloalkylene, cycloalkenyl, cycloalkenylene, cycloalkynyl, and cycloalkynylene, and the like.
The terms "cycloalkyl" or "cycloalkyl group" mean a stable aliphatic saturated 3- to 15- membered monocyclic or polycyclic monovalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornane, adamantyl, tetrahydronaphthyl (tetralin), 1-decalinyl, bicyclo[2.2.2]octanyl, 1 -methylcyclopropyl, 2-methylcyclopentyl, 2- methylcyclooctyl, and the like.
The terms "cycloalkenyl" or "cycloalkenyl group" mean a stable aliphatic 5- to 15-membered monocyclic or polycyclic monovalent radical having at least one carbon-carbon double bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkenyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkenyl groups include cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, cyclononenyl, cyclodecenyl, norbornenyl, 2-methylcyclopentenyl, 2-methylcyclooctenyl, and the like.
The terms "cycloalkynyl" or "cycloalkynyl group" mean a stable aliphatic 8- to 15-membered monocyclic or polycyclic monovalent radical having at least one carbon-carbon triple bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 8- to 10-membered monocyclic or 12- to 15-membered bicyclic ring. Unless otherwise specified, the cycloalkynyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkynyl groups include, cyclooctynyl, cyclononynyl, cyclodecynyl, 2-methylcyclooctynyl, and the like.
The terms "cycloalkylene" or "cycloalkylene group" mean a stable saturated aliphatic 3- to 15-membered monocyclic or polycyclic divalent radical consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkyl ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkylene groups include cyclopentylene, and the like.
The terms "cycloalkenylene" or "cycloalkenylene group" mean a stable aliphatic 5- to 15- membered monocyclic or polycyclic divalent radical having at least one carbon-carbon double bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring. Unless otherwise specified, the cycloalkenylene ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkenylene groups include cyclopentenylene, cyclohexenylene, cycloheptenylene, cyclooctenylene, cyclononenylene, cyclodecenylene, norbornenylene, 2-methylcyclopentenylene, 2- methylcyclooctenylene, and the like. The terms "cycloalkynylene" or "cycloalkynylene group" mean a stable aliphatic 8- to 15- membered monocyclic or polycyclic divalent radical having at least one carbon-carbon triple bond and consisting solely of carbon and hydrogen atoms which may comprise one or more fused or bridged ring(s), preferably a 8- to 10-membered monocyclic or 12- to 15-membered bicyclic ring. Unless otherwise specified, the cycloalkynylene ring may be attached at any carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary cycloalkynylene groups include cyclooctynylene, cyclononynylene, cyclodecynylene, 2-methylcyclooctynylene, and the like.
The terms "aryl" or "aryl group" mean an aromatic carbocyclic monovalent or divalent radical of from 6 to 14 carbon atoms having a single ring (e.g., phenyl or phenylene) or multiple condensed rings (e.g., naphthyl or anthranyl). Unless otherwise specified, the aryl ring may be attached at any suitable carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable carbon atom which results in a stable structure. Exemplary aryl groups include phenyl, naphthyl, dihydronaphthyl, tetrahydronaphthyl, anthryl, phenanthryl, indanyl, indenyl, biphenyl, and the like. It may be abbreviated "Ar".
The terms "heteroaryl" or "heteroaryl group" mean a stable aromatic 5- to 14-membered, monocyclic or polycyclic monovalent or divalent radical which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic radical, having from one to four heteroatoms in the ring(s) independently selected from nitrogen, oxygen, and sulfur, wherein any sulfur heteroatoms may optionally be oxidized and any nitrogen heteroatom may optionally be oxidized or be quaternized. Unless otherwise specified, the heteroaryl ring may be attached at any suitable heteroatom or carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable heteroatom or carbon atom which results in a stable structure. Exemplary and preferred heteroaryls include furanyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, indolyl, azaindolyl, dihydroindolyl, isoindolyl, benzofuranyl, dihydrobenzofuranyl, benzopyranyl, dihydrobenzopyranyl, benzpyrrolyl, benzodioxepinyl, dihydrobenzodioxepinyl, benzothienyl, dihydrobenzothienyl, indazolyl, benzimidazolyl, benzthiazolyl, benzoxazolyl, benzisoxazolyl, benzpyrazolyl, purinyl, quinolizinyl, quinolinyl, dihydroquinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, isoquinolinyl, dihydroisoquinolinyl, tetrahydroisoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, and phenoxazinyl, and the like.
The terms "heterocycle", "heterocycle group", "heterocyclyl", or "heterocyclyl group" mean a stable non-aromatic 5- to 14-membered monocyclic or polycyclic, monovalent or divalent, ring which may comprise one or more fused or bridged ring(s), preferably a 5- to 7-membered monocyclic or 7- to 10-membered bicyclic ring, having from one to three heteroatoms in the ring(s) independently selected from nitrogen, oxygen, and sulfur, wherein any sulfur heteroatoms may optionally be oxidized and any nitrogen heteroatom may optionally be oxidized or be quaternized. Unless otherwise specified, the heterocyclyl ring may be attached at any suitable heteroatom or carbon atom which results in a stable structure and, if substituted, may be substituted at any suitable heteroatom or carbon atom which results in a stable structure. Exemplary and preferred heterocycles include pyrrolinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrofuranyl, hexahydropyrimidinyl, hexahydropyridazinyl, and the like.
The term "compounds of the invention" and equivalent expressions are meant to embrace compounds of Formula (I) as herein described, including the tautomers, the prodrugs, the salts, particularly the pharmaceutically acceptable salts, and the solvates and hydrates thereof, where the context so permits. In general and preferably, the compounds of the invention and the formulas designating the compounds of the invention are understood to only include the stable compounds thereof and exclude unstable compounds, even if an unstable compound might be considered to be literally embraced by the compound formula. Similarly, reference to intermediates, whether or not they themselves are claimed, is meant to embrace their salts and solvates, where the context so permits. For the sake of clarity, particular instances when the context so permits are sometimes indicated in the text, but these instances are purely illustrative and it is not intended to exclude other instances when the context so permits. The terms "optional" or "optionally" mean that the subsequently described event or circumstances may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
The terms "stable compound" or "stable structure" mean a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic or diagnostic agent. For example, a compound which would have a "dangling valency" or is a carbanion is not a compound contemplated by the invention.
The term "substituted" means that any one or more hydrogens on an atom of a group or moiety, whether specifically designated or not, is replaced with a selection from the indicated group of substituents, provided that the atom's normal valency is not exceeded and that the substitution results in a stable compound. If a bond to a substituent is shown to cross the bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom via which such substituent is bonded to the rest of the compound, then such substituent may be bonded via any atom in such substituent. For example, when the substituent is piperazinyl, piperidinyl, or tetrazolyl, unless specified otherwise, such piperazinyl, piperidinyl, or tetrazolyl group may be bonded to the rest of the compound of the invention via any atom in such piperazinyl, piperidinyl, or tetrazolyl group. Generally, when any substituent or group occurs more than one time in any constituent or compound, its definition on each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0 to 2 R5, then such group is optionally substituted with up to two R5 groups and R5 at each occurrence is selected independently from the defined list of possible R5. Such combinations of substituents and/or variables, however, are permissible only if such combinations result in stable compounds.
In a specific embodiment, the term "about" or "approximately" means within 20%, preferably within 10%, and more preferably within 5% of a given value or range. The yield of each of the reactions described herein is expressed as a percentage of the theoretical yield.
B. Salt, Prodrug, Derivative, and Solvate Terms and Conventions
The terms "prodrug" or "prodrug derivative" mean a covalently-bonded derivative or carrier of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s). In general, such prodrugs have metabolically cleavable groups and are rapidly transformed in vivo to yield the parent compound, for example, by hydrolysis in blood, and generally include esters and amide analogs of the parent compounds. The prodrug is formulated with the objectives of improved chemical stability, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity). In general, prodrugs themselves have weak or no biological activity and are stable under ordinary conditions. Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5: "Design and Applications of Prodrugs"; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery. K.B. Sloan (ed.), Marcel Dekker, 1998; Methods in Enzymology, K. Widder et al. (eds.), Vol. 42, Academic Press, 1985, particularly pp. 309- 396; Burger's Medicinal Chemistry and Drug Discovery, 5th Ed., M. Wolff (ed.), John Wiley & Sons, 1995, particularly Vol. 1 and pp. 172-178 and pp. 949-982; Pro-Drugs as Novel Delivery Systems, T. Higuchi and V. Stella (eds.), Am. Chem. Soc, 1975; and Bioreversible Carriers in Drug Design, E.B. Roche (ed.), Elsevier, 1987, each of which is incorporated herein by reference in their entireties.
The term "pharmaceutically acceptable prodrug" as used herein means a prodrug of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible. The term "salt" means an ionic form of the parent compound or the product of the reaction between the parent compound with a suitable acid or base to make the acid salt or base salt of the parent compound. Salts of the compounds of the present invention can be synthesized from the parent compounds which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts are prepared by reacting the free base or acid parent compound with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base in a suitable solvent or various combinations of solvents.
The term "pharmaceutically acceptable salt" means a salt of a compound of the invention which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use. The term includes pharmaceutically- acceptable acid addition salts and pharmaceutically-acceptable base addition salts. As the compounds of the present invention are useful in both free base and salt form, in practice, the use of the salt form amounts to use of the base form. Lists of suitable salts are found in, e.g., S.M. Birge et at, J. Pharm. ScL, 1977, 66, pp. 1-19, which is hereby incorporated by reference in its entirety.
The term "pharmaceutically-acceptable acid addition salt" means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trichloroacetic acid, trifluoroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 2- acetoxybenzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, hemisulfic acid, heptanoic acid, hexanoic acid, formic acid, fumaric acid, 2- hydroxyethanesulfonic acid (isethionic acid), lactic acid, maleic acid, hydroxymaleic acid, malic acid, malonic acid, mandelic acid, mesitylenesulfonic acid, methanesulfonic acid, naphthalenesulfonic acid, nicotinic acid, 2-naphthalenesulfonic acid, oxalic acid, pamoic acid, pectinic acid, phenylacetic acid, 3-phenylpropionic acid, picric acid, pivalic acid, propionic acid, pyruvic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, sulfanilic acid, tartaric acid, p-toluenesulfonic acid, undecanoic acid, and the like.
The term "pharmaceutically-acceptable base addition salt" means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a metal cation such as sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically-acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, quaternary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion-exchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2- dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tetramethylammonium compounds, tetraethylammonium compounds, pyridine, N,N-dimethylaniline, N- methylpiperidine, N-methylmorpholine, dicyclohexylamine, dibenzylamine, N5N- dibenzylphenethylamine, 1-ephenamine, N,N'-dibenzylethylenediamine, polyamine resins, and the like. Particularly preferred organic nontoxic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
The term "solvate" means a physical association of a compound with one or more solvent molecules or a complex of variable stoichiometry formed by a solute (for example, a compound of Formula (I)) and a solvent, for example, water, ethanol, or acetic acid. This physical association may involve varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. In general, the solvents selected do not interfere with the biological activity of the solute. Solvates encompasses both solution-phase and isolatable solvates. Representative solvates include hydrates, ethanolates, methanolates, and the like.
The term "hydrate" means a solvate wherein the solvent molecule(s) is/are H2O.
The compounds of the present invention as discussed below include the free base or acid thereof, their salts, solvates, and prodrugs and may include oxidized sulfur atoms or quaternized nitrogen atoms in their structure, although not explicitly stated or shown, particularly the pharmaceutically acceptable forms thereof. Such forms, particularly the pharmaceutically acceptable forms, are intended to be embraced by the appended claims.
C. Isomer Terms and Conventions
The term "isomers" means compounds having the same number and kind of atoms, and hence the same molecular weight, but differing with respect to the arrangement or configuration of their atoms in space. The term includes stereoisomers and geometric isomers.
The terms "stereoisomer" or "optical isomer" means a stable isomer that has at least one chiral atom or restricted rotation giving rise to perpendicular dissymmetric planes (e.g., certain biphenyls, allenes, and spiro compounds) and can rotate plane-polarized light. Because asymmetric centers and other chemical structure exist in the compounds of the invention which may give rise to stereoisomerism, the invention contemplates stereoisomers and mixtures thereof. The compounds of the invention and their salts include asymmetric carbon atoms and may therefore exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers. Typically, such compounds will be prepared as a racemic mixture. If desired, however, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. As discussed in more detail below, individual stereoisomers of compounds are prepared by synthesis from optically active starting materials containing the desired chiral centers or by preparation of mixtures of enantiomeric products followed by separation or resolution, such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, use of chiral resolving agents, or direct separation of the enantiomers on chiral chromatographic columns. Starting compounds of particular stereochemistry are either commercially available or are made by the methods described below and resolved by techniques well-known in the art.
The term "enantiomers" means a pair of stereoisomers that are non-superimposable mirror images of each other.
The terms "diastereoisomers" or "diastereomers" mean stereoisomers which are not mirror images of each other.
The terms "racemic mixture" or "racemate" mean a mixture containing equal parts of individual enantiomers.
The term "non-racemic mixture" means a mixture containing unequal parts of individual enantiomers.
The term "geometrical isomer" means a stable isomer which results from restricted freedom of rotation about double bonds (e.g., cώ-2-butene and trans-2-butQm) or in a cyclic structure (e.g., czs-l,3-dichlorocyclobutane and £rø«s-l,3-dichlorocyclobutane). Because carbon- carbon double (olefinic) bonds, C=N double bonds, cyclic structures, and the like may be present in the compounds of the invention, the invention contemplates each of the various stable geometric isomers and mixtures thereof resulting from the arrangement of substituents around these double bonds and in these cyclic structures. The substituents and the isomers are designated using the cisltrans convention or using the E ox Z system, wherein the term "E" means higher order substituents on opposite sides of the double bond, and the term "Z" means higher order substituents on the same side of the double bond. A thorough discussion of E and Z isomerism is provided in J. March, Advanced Organic Chemistry: Reactions. Mechanisms, and Structure, 4th ed., John Wiley & Sons, 1992, which is hereby incorporated by reference in its entirety. Several of the following examples represent single E isomers, single Z isomers, and mixtures OΪ EIZ isomers. Determination of the E and Z isomers can be done by analytical methods such as x-ray crystallography, 1H NMR, and 13C NMR. Some of the compounds of the invention can exist in more than one tautomeric form. As mentioned above, the compounds of the invention include all such tautomers.
It is well-known in the art that the biological and pharmacological activity of a compound is sensitive to the stereochemistry of the compound. Thus, for example, enantiomers often exhibit strikingly different biological activity including differences in pharmacokinetic properties, including metabolism, protein binding, and the like, and pharmacological properties, including the type of activity displayed, the degree of activity, toxicity, and the like. Thus, one skilled in the art will appreciate that one enantiomer may be more active or may exhibit beneficial effects when enriched relative to the other enantiomer or when separated from the other enantiomer. Additionally, one skilled in the art would know how to separate, enrich, or selectively prepare the enantiomers of the compounds of the invention from this disclosure and the knowledge of the prior art.
Thus, although the racemic form of drug may be used, it is often less effective than administering an equal amount of enantiomerically pure drug; indeed, in some cases, one enantiomer may be pharmacologically inactive and would merely serve as a simple diluent. For example, although ibuprofen had been previously administered as a racemate, it has been shown that only the 5*-isomer of ibuprofen is effective as an anti-inflammatory agent (in the case of ibuprofen, however, although the R -isomer is inactive, it is converted in vivo to the S- isomer, thus, the rapidity of action of the racemic form of the drug is less than that of the pure .S-isomer). Furthermore, the pharmacological activities of enantiomers may have distinct biological activity. For example, ^-penicillamine is a therapeutic agent for chronic arthritis, while ^-penicillamine is toxic. Indeed, some purified enantiomers have advantages over the racemates, as it has been reported that purified individual isomers have faster transdermal penetration rates compared to the racemic mixture. See U.S. Patent Nos. 5,114,946 and 4,818,541.
Thus, if one enantiomer is pharmacologically more active, less toxic, or has a preferred disposition in the body than the other enantiomer, it would be therapeutically more beneficial to administer that enantiomer preferentially. In this way, the patient undergoing treatment would be exposed to a lower total dose of the drug and to a lower dose of an enantiomer that is possibly toxic or an inhibitor of the other enantiomer.
Preparation of pure enantiomers or mixtures of desired enantiomeric excess (ee) or enantiomeric purity are accomplished by one or more of the many methods of (a) separation or resolution of enantiomers, or (b) enantioselective synthesis known to those of skill in the art, or a combination thereof. These resolution methods generally rely on chiral recognition and include, for example, chromatography using chiral stationary phases, enantioselective host-guest complexation, resolution or synthesis using chiral auxiliaries, enantioselective synthesis, enzymatic and nonenzymatic kinetic resolution, or spontaneous enantioselective crystallization. Such methods are disclosed generally in Chiral Separation Techniques: A Practical Approach (2nd Ed.), G. Subramanian (ed.), Wiley- VCH, 2000; T.E. Beesley and R.P.W. Scott, Chiral Chromatography, John Wiley & Sons, 1999; and Satinder Ahuja, Chiral Separations by Chromatography, Am. Chem. Soc, 2000. Furthermore, there are equally well-known methods for the quantitation of enantiomeric excess or purity, for example, GC, HPLC, CE, or NMR, and assignment of absolute configuration and conformation, for example, CD ORD, X-ray crystallography, or NMR.
In general, all tautomeric forms and isomeric forms and mixtures, whether individual geometric isomers or stereoisomers or racemic or non-racemic mixtures, of a chemical structure or compound is intended, unless the specific stereochemistry or isomeric form is specifically indicated in the compound name or structure.
D. Pharmaceutical Administration and Diagnostic and Treatment Terms and Conventions
The term "patient" includes both human and non-human mammals.
The term "effective amount" means an amount of a compound according to the invention which, in the context of which it is administered or used, is sufficient to achieve the desired effect or result. Depending on the context, the term effective amount may include or be synonymous with a pharmaceutically effective amount or a diagnostically effective amount. The terms "pharmaceutically effective amount" or "therapeutically effective amount" means an amount of a compound according to the invention which, when administered to a patient in need thereof, is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue, system, or patient that is sought by a researcher or clinician. The amount of a compound of according to the invention which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the invention, and the age, body weight, general health, sex, and diet of the patient. Such a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure.
The term "diagnostically effective amount" means an amount of a compound according to the invention which, when used in a diagnostic method, apparatus, or assay, is sufficient to achieve the desired diagnostic effect or the desired biological activity necessary for the diagnostic method, apparatus, or assay. Such an amount would be sufficient to elicit the biological or medical response in a diagnostic method, apparatus, or assay, which may include a biological or medical response in a patient or in a in vitro or in vivo tissue or system, that is sought by a researcher or clinician. The amount of a compound according to the invention which constitutes a diagnostically effective amount will vary depending on such factors as the compound and its biological activity, the diagnostic method, apparatus, or assay used, the composition used for administration, the time of administration, the route of administration, the rate of excretion of the compound, the duration of administration, drugs and other compounds used in combination with or coincidentally with the compounds of the invention, and, if a patient is the subject of the diagnostic administration, the age, body weight, general health, sex, and diet of the patient. Such a diagnostically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure. The term "modulate" means the ability of a compound to alter the function of the glucocorticoid receptor by, for example, binding to and stimulating or inhibiting the glucocorticoid receptor functional responses.
The term "modulator" in the context of describing compounds according to the invention means a compound that modulates the glucocorticoid receptor function. As such, modulators include, but are not limited to, agonists, partial agonists, antagonists, and partial antagonists.
The term "agonist" in the context of describing compounds according to the invention means a compound that, when bound to the glucocorticoid receptor, enhances or increases the glucocorticoid receptor function. As such, agonists include partial agonists and full agonists.
The term "full agonist" in the context of describing compounds according to the invention means a compound that evokes the maximal stimulatory response from the glucocorticoid receptor, even when there are spare (unoccupied) glucocorticoid receptors present.
The term "partial agonist" in the context of describing compounds according to the invention means a compound that is unable to evoke the maximal stimulatory response from the glucocorticoid receptor, even at concentrations sufficient to saturate the glucocorticoid receptors present.
The term "antagonist" in the context of describing compounds according to the invention means a compound that directly or indirectly inhibits or suppresses the glucocorticoid receptor function. As such, antagonists include partial antagonists and full antagonists.
The term "full antagonist" in the context of describing compounds according to the invention means a compound that evokes the maximal inhibitory response from the glucocorticoid receptor, even when there are spare (unoccupied) glucocorticoid receptors present.
The term "partial antagonist" in the context of describing compounds according to the invention means a compound that is unable to evoke the maximal inhibitory response from the glucocorticoid receptor, even at concentrations sufficient to saturate the glucocorticoid receptors present.
The terms "treating" or "treatment" mean the treatment of a disease-state in a patient, and include:
(i) preventing the disease-state from occurring in a patient, in particular, when such patient is genetically or otherwise predisposed to the disease-state but has not yet been diagnosed as having it;
(ii) inhibiting or ameliorating the disease-state in a patient, i.e., arresting or slowing its development; or
(iii) relieving the disease-state in a patient, i.e., causing regression or cure of the disease- state.
General Synthetic Methods for Making Compounds of Formula (IA) and Formula (IB) The invention also provides processes for making compounds of Formula (IA) and Formula (IB). In all schemes, unless specified otherwise, R1, R2, R3, R4, R5, R6, X, and R7 in the formulas below shall have the meaning of R1, R2, R3, R4, R5, R6, X, and R7, in the Formula (IA) of the invention described hereinabove; and R1, R2, R3, R4, R5, R6, X, R7, and R8 in the formulas below shall have the meaning of R1, R2, R3, R4, R5, R6, X, R7, and R8, in the Formula (IB) of the invention described hereinabove. Intermediates used in the preparation of compounds of the invention are either commercially available or readily prepared by methods known to those skilled in the art.
Optimum reaction conditions and reaction times may vary depending on the particular reactants used. Unless otherwise specified, solvents, temperatures, pressures, and other reaction conditions may be readily selected by one of ordinary skill in the art. Specific procedures are provided in the Synthetic Examples section. Typically, reaction progress may be monitored by thin layer chromatography (TLC), if desired, and intermediates and products may be purified by chromatography on silica gel and/or by recrystallization.
Compounds of Formula (IA) where R8 is an optionally substituted aryl group may be prepared by the method outlined in Scheme I. Compounds of Formula (IA) and (IB) where R6 and R8, in Formula (IA) and (IB) respectively, are optionally substituted heteroaryl group may be prepared by the method outlined in Scheme I.
Figure imgf000063_0001
IV V Vl
Figure imgf000063_0002
VII IA
Scheme I
As illustrated in Scheme I, an amino acid where R' is H or an amino acid ester where R' is Me or Et (IV) is reacted with a sulfonyl chloride (V) in a suitable solvent, such as THF, in the presence of a suitable base, such as NaH, to provide sulfonamide (VI). Reduction of sulfonamide (VI) with a suitable reducing agent, such as lithium aluminum hydride, provides alcohol (VII). Ring closure by methods known in the art, for example, reacting the alcohol with a sulfonyl chloride such a /?βrø-toluenesulfonyl chloride in the presence of a suitable base, such as sodium hydride, provides the aziridine (II). The aziridine (II) is reacted with a reagent of R6X-M of Formula (III) where X is sulfur, oxygen, or NR7 and M is Na, K or Li derived from R6X-H were X is oxygen using a suitable base such as sodium hydride in a suitable solvent such as DMF or DMSO; and from R6X-H were X is sulfur using a suitable base such as sodium hydride in a suitable solvent such as DMF or DMSO, or 2-tert- butylimino-2-diehylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine on polystyrene in a suitable solvent such as acetonitrile and from R6X-H were X is NR7 using sodium bis(trimethylsilyl)amide in a suitable solvent such a DMSO or DMF. Alternatively, the aziridine (II) is reacted with a reagent R6X-M of Formula (III) where X is NR7 or sulfur and M is hydrogen in a suitable solvent such as THF under thermal conditions to form the compound of Formula (IA). The racemic and chiral amino acids and amino acid esters (IV) as well as the sulfonyl chlorides R SO2CI (V) are either commercially available or may be readily prepared by methods known to those skilled in the art. Hence, enantiomerically enriched compounds of Formula (IA) may be prepared by using chiral starting materials. For example, a method of preparing 1 , 1 , 1 -trifluoroalanine is given in V.A. Soloshonok et ah, Tetrahedron, 1997, 53, 8307.
Another approach that may be used to obtain compounds of Formula (IA) is illustrated in Scheme II.
Figure imgf000064_0001
Figure imgf000064_0002
Scheme II
As illustrated in Scheme II, an amine of Formula (VIII) is reacted with a sulfonyl chloride of Formula (V) in a suitable solvent such as pyridine in to form an sulfonamide of Formula (IX). The thiol of Formula (IX) is reacted with an oxonium salt such as trimethyloxonium tetrafluoroborate in a suitable solvent such as dichloromethane in to form a sulfonium salt of Formula (X). Cyclization the sulfonium salt of Formula (X) in a suitable solvent such as tetrahydrofuran in the presence of a suitable base such as sodium hydride to form an aziridine of Formula (II) where R3, R4, and R5 are each H which may be converted to a compound of Formula (IA) as in Scheme I.
Chiral α-fluorinated amines of Formula (VIII) as well as the sulfonyl chlorides R2SC^Cl (V) are either commercially available or may be readily prepared by methods known to those skilled in the art. Hence, enantiomerically enriched compounds of Formula (IA) may be prepared by using chiral starting materials. For example, a method of preparing chiral and racemic α-fluorinated amines of Formula (VIII) is given in P. Bravo et ah, J. Org. Chem., 1996, 61, 3375. A method of preparing chiral aziridines is given in A. Toshimitsu et ah, J. Chem. Soc, Chem. Commun, 1992, 284.
Compounds of Formula (IB) may be prepared using a general procedure outlined in Scheme III. This general procedure is suitable for a variety of R2 and R3 (for this example, R3 is ethyl) where R8 is an optionally substituted heteroaryl group and X is sulfur, oxygen, or nitrogen (for this example, X is nitrogen and R7 is H).
* R»-NH2
Figure imgf000065_0001
XII XIII XIX
Figure imgf000065_0002
IB
Scheme III
As illustrated in Scheme III, the optionally substituted amino alcohol (XII) bearing R3 is reacted with a sulfonyl chloride (V) bearing R in a suitable solvent such as tetrahydrofuran in the presence of a base such as sodium hydride or in dichloromethane in the presence of pyridine followed cyclization with a suitable base such as aqueous potassium hydroxide to form an aziridine of Formula (XIII). The aziridine (XIII) is reacted with a suitable organometallic reagent R8X-M where X is sulfur, oxygen, or NR7 and M is Li, Na, or K derived from R8X-H were X is oxygen using a suitable base such as sodium hydride in a suitable solvent, such as DMF or DMSO; and from R8X-H were X is sulfur using a suitable base such as sodium hydride in a suitable solvent, such as DMF or DMSO, or 2-tert- butylimino-2-diethylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine on polystyrene in a suitable solvent, such as acetonitrile, and from R8X-H where X is NR7 using sodium bis(trimethylsilyl)amide in a suitable solvent, such a DMSO or DMF. Alternatively, the aziridine (XIII) is reacted with a reagent R8X-M where X is NR7 or sulfur and M is hydrogen in a suitable solvent, such as THF, under thermal conditions with or without an additive, such as lithium perchlorate in acetonitrile or β-cyclodextrin hydrate in methanol, to form the compound of Formula (IB).
Racemic and chiral aminoalcohols (XII) are either commercially available or may be readily prepared by methods known to those skilled in the art. The sulfonyl chlorides R2SC^Cl (V) are either commercially available or may be readily prepared by methods known to those skilled in the art. For example, a general method of preparing sulfonyl chloride from anilines is given in R.V. Hoffman, Org. Synth. 1981, 60, 121. Aziridines (XIII) may also be commercially available or prepared from aminoalcohols by methods known to those skilled in the art. For example, methods of preparing aziridines are given in M.B. Berry and D. Craig, Synlett 1992, 41; J. Farras, et al. Tetrahedron 2001, 57, 7665; W. Oppolzer, et al. Helveica Chimica Acta 2001, 84, 141 and C. Moberg, et al. Tetrahedron Asymmetry, 1997, 15, 2655.
Another approach that may be used to obtain compounds of Formula (IA) or (IB) suitable for aa vvaarriieettyy ooff RR22 ggrroouuppss iiss iillllustrated in Scheme IV for this example R3 is ethyl to provide a compound of Formula (IB).
Figure imgf000066_0001
Figure imgf000066_0002
IB Scheme IV
As illustrated in Scheme IV, a compound of Formula (IB) bearing R8, bearing R3 (for this example, R3 is ethyl), bearing XR7 (for this example, X is nitrogen and R7 is H), and an R2 (for this example, R2 is o-nitropheny) is reacted with thiophenol and a suitable base such as potassium carbonate in a suitable solvent such as DMF to provide the amine (XX). The aminoethyl compound of Formula (XX) is sulfonated with a sulfonyl halide (for this example, R2 is 2,4-dichloro-6-aminophenyl) of Formula (XXI) in the presence of a suitable base such as triethyamine or pyridine in a suitable solvent such as dichloromethane to form the compound of Formula (IB) where R is not o-nitrophenyl.
Another approach that may be used to obtain compounds of Formula (IA) or (IB), where X is NR7, suitable for a variety of R7 groups is illustrated in Scheme V.
→"j
Figure imgf000067_0001
XXII V XXIII XXIV
Figure imgf000067_0002
XXV IB
Scheme V
As illustrated in Scheme V, the optionally substituted amino acid of Formula (XXII) useful to provide compounds of Formula (IB) bearing R3 (for this example, R3 is isopropyl) is reacted with a sulfonyl chloride (VII) in a suitable solvent, such as acetone-water, in the presence of a suitable base, such as NaOH to provide sulfonamide of Formula (XXIII). A coupling reaction of a carboxylic acid of Formula (XXIII) with an aryl amine (in this example N-ethylaniline) using an activating agent, such as thiophosgene, in a suitable solvent, such as chloroform, in the presence of DMF to form an amide of Formula (XXV). Reduction of an amide of Formula (XXV) with a suitable reducing agent, such as borane in a suitable solvent, such as THF, provides compounds of Formula (IB). In a similar fashion, compounds of (IA) can be obtained from 1 ,1 ,1-trifluoroalanine.
In order that this invention be more fully understood, the following examples are set forth. These examples are for the purpose of illustrating embodiments of this invention, and are not to be construed as limiting the scope of the invention in any way since, as recognized by one skilled in the art, particular reagents or conditions could be modified as needed for individual compounds. Starting materials used are either commercially available or easily prepared from commercially available materials by those skilled in the art.
Experimental Examples Example 1: iV-{(S)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6- trimethylbenzenesulfonamide
Figure imgf000068_0001
A mixture of 120 mg (0.47 mmol) of (S)-2-ethyl-l-(2,4,6-trimethylbenzenesulfonyl)- aziridine, 62.6 mg (0.47 mmol) of 4-aminoindole, and 177.4 mg (0.16 mmol) of β- cyclodextrin hydrate in 1.5 mL of methanol in a sealed tube was warmed at 65°C in a microwave. After 24 hours, the mixture was concentrated in vacuo and purified first by chromatography on silica gel eluting with EtOAc-hexanes (0-40% gradient), and second by reversed phase HPLC to afford the title compound as the trifluoroacetic acid salt. LCMS M+ = 386.24.
Example 2: iV-((S)-l-{[l-(4-Fluorophenyl)-lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide
Figure imgf000068_0002
A mixture of 120.0 mg (0.47 mmol) of (5)-2-ethyl-l-(2,4,6-trimethylbenzenesulfonyl)- aziridine, 107.6 mg (0.47 mmol) of l-(4-fluorophenyl)-lH-indazol-5-ylamine, and 177.4 mg (0.16 mmol) of β-cyclodextrin hydrate in 1.5 mL of methanol in a sealed tube was warmed at 65°C in a microwave. After 24 hours, the mixture was concentrated in vacuo and purified first by chromatography on silica gel eluting with EtOAc-hexanes (0-50% gradient), and second by reversed phase HPLC to afford the title compound as the trifluoroacetic acid salt. LCMS M+ = 481.27.
Example 3: 2-Amino-4,6-dichloro-ΛL((S)-l-{[l-(4-fluorophenyl)-lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide
Figure imgf000069_0001
To a mixture of 2.6 g (29.1 mmol) of (5)-(+)-2-amino- 1 -butanol (98%) in 50 mL of dichloromethane and 20 mL of pyridine was added 16.4 g (74.0 mmol) of o- nitrobenzenesulfonyl chloride in several portions. The mixture stirred for over night and was then diluted with 100 mL of ether and washed with six 50 mL portions of 1 N aqueous HCl until the aqueous layer was acidic. The ether layer was then diluted with 40 mL of 2 N aqueous KOΗ and stirred for 6 hours. The basic aqueous layers was separated and the ether layer were washed with two 40 mL portions of 1 N aqueous KOH, 20 mL of brine, three 20 mL portions of saturated aqueous ammonium chloride, dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was passed thru a pad of silica gel eluting with dichloromethane-hexanes (25:75) and then ethyl acetate-hexanes (3:7) to afford 2.05 g (52%) of (5)-2-ethyl-l-(2-nitrobenzenesulfonyl)aziridine. Mixed fractions from the column were chromatographed on silica gel using dichloromethane-hexanes (2:8) and then ethyl acetate- hexanes (4:6) to afford an additional 1.87 g of (5)-2-ethyl- 1 -(2-nitrobenzenesulfonyl)aziridine which solidified upon standing.
A mixture of 256.3 mg (1.0 mmol) of (5)-2-ethyl-l-(2-nitrobenzenesulfonyl)aziridine, 227.3 mg (1.0 mmol) of l-(4-fluorophenyl)-lH-indazol-5-ylamine, and 374.5 mg (0.33 mmol) of β- cyclodextrin hydrate in 3.0 mL of methanol in a sealed tube was warmed at 65°C in a microwave. After 18 hours, the mixture was concentrated in vacuo and purified by chromatography on silica gel eluting with EtOAc-hexanes (10-100% gradient) to afford 305 mg (63%) of N-((5)-l-{[l-(4-fluorophenyl)-lH-indazol-5-ylamino]methyl}propyl)-2- nitrobenzenesulfonamide. LCMS M+ = 484.35.
To a solution of 250.0 mg (0.52 mmol) of N-((S)-l-{[l-(4-fluorophenyl)-lH-indazol-5- ylamino]methyl}propyl)-2-nitrobenzenesulfonamide in 10 mL of DMF was added 714 mg (5.17 mmol) Of K2CO3 followed by 106 μL (1.03 mmol) of thiophenol. After 18 hours, the mixture was diluted with 30 mL of diethyl ether, made acidic with 1 N aqueous HCl, and the acidic aqueous layer was separated. The organic layer was extracted with two 30 mL portions of IN aqueous HCl. The combined acidic aqueous layers were washed with three 30 mL portions of ether, made basic with potassium carbonate and extracted with three 30 mL portions of ethyl acetate. The combined organic layers were washed with two 30 mL portions of brine, dried over magnesium sulfate, filtered, and concentrated in vacuo provide 150 mg (97%) of (5)-N1-[l-(4-fluorophenyl)-lH-indazol-5-yl]butane-l,2-diamine as an oil which was used without further purification. LCMS M+ = 299.34.
A mixture of 150 mg (0.50 mmol) of (5)-N1-[l-(4-fluorophenyl)-lH-indazol-5-yl]butane-l,2- diamine and 131 mg (0.50 mmol) of 2-amino-4,6-dichlorosulfonyl chloride in 10 mL of dichloromethane was treated with 210 μL (1.51 mmol) of triethylamine. After stirring at room temperature for 50 hours, the mixture was concentrated and dissolved in 30 mL of EtOAc and washed with three 30 mL portions of saturated aqueous sodium bicarbonate, 30 mL of brine, dried over magnesium sulfate, filtered, and concentrated. The mixture was purified by first silica gel chromatography eluting with ethyl acetate-hexanes (20-100% gradient) to afford 208 mg (79%) of the title compound. LCMS M+ = 522.35. This material was further purified by Prep-TLC chromatography (20% EtOAc/hexanes, developed 3 times) and then by RP-HPLC. The product rich fractions were pooled and concentrated to remove acetonitrile. The mixture was treated with 10 mL of dichloromethane and diluted with 1 N aqueous NaOH to pH 10. The aqueous phase was separated and extracted with two 10 mL portions of dichloromethane. The combined organic layers were washed with two 10 mL portions of brine, dried over magnesium sulfate, filtered, and concentrated in vacuo to afford 69.7 mg (26%) of the title compound as a tan foam. LCMS M+ = 522.33.
Example 4: 2,4,6-Trimethyl-iV-[2,2,2-trifluoro-l-(quinolin-4- yloxymethyl)ethyl]benzenesulfonamide
Figure imgf000071_0001
To a solution of 45 mg (0.31 mmol) of 4-hydroxyquinoline in 2 mL of DMF was added 33.0 mg (0.83 mmol) of sodium hydride (60% in mineral oil). The mixture stirred until hydrogen evolution ceased and then 75.0 mg (0.26 mmol) of 2-trifluoromethyl- 1 -(2,4,6- trimethylbenzenesulfonyl)aziridine was added. The reaction was monitored by TLC (MeOH- CH2CI2). After 1 hour, the mixture was quenched with 10 mL of saturated aqueous ammonium chloride and extracted with three 10 mL portions of ethyl acetate. The combined organic layers were washed with three 10 mL portions of brine, dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was triturated with ether and crystallized from dichloromethane-methanol-ether to afford a 1.0:0.1 mixture of two compounds as determined by IH NMR (400 MHz, DMSO-d6). The solid and the filtrates were chromatographed on silica gel (prep plates, 1 mm, 10% methanol-dichloromethane). The first eluting band was triturated with ether to afford 15 mg (13%) of the title compound: m.p. 211- 212°C, IR spectrum lacks a carbonyl stretch and LCMS M+ = 439.54. And a second band which was triturated with ether to afford 60 mg (53%) of the N-linked quinoline: m.p. 276- 279°C, IR (KBr) 1622 cm"1, LCMS M+ = 439.54.
Example 5: 2,4,6-Trimethyl-iV-[2,2,2-trifluoro-l-(quinolin-5- ylaminomethyl)ethyl]benzenesulfonamide
Figure imgf000072_0001
A solution of 0.12 mmol of 2-trifluoromethyl-l-(2,4,6-trimethylbenzenesulfonyl)aziridine in 0.8 mL DMSO was added to a vial containing 0.12 mmol of quinolin-5-ylamine. 120 mL of a 1.0 M solution of sodium bis(trimethylsilyl)amide) (NaHMDS) in tetrahydrofuran was then added. The vial was placed on an orbital shaker overnight. 20 mL of water was added and the vial was placed back on the shaker for 1 hour. The contents of each vial were transferred to a microtube plate. The compound was purified by mass triggered prep LC and all fractions were evaporated to afford the title compound and was analyzed by NMR and HPLC.
Example 6: 2,4,6-Trimethyl-iV-[2,2,2-trifluoro-l-(4-thiophen-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide
Figure imgf000072_0002
35 mg (0.03 mmol) of β-cyclocodextrin was added to 4-thiophen-2-ylpyrimidine-2-thiol in a vial using a resin scoop. 0.1 mmol of 2-trifluoromethyl- 1 -(2,4,6- trimethylbenzenesulfonyl)aziridine in 0.90 mL of MeOH was added. The vials were placed on an orbital shaker at 500C. After 36 hours, the reaction was cooled to room temperature, filtered and washed with dichloroethane. Plate was evaporated and the reactions were transferred to microtube plate with three 300 μL portions of DMSO. The compound was purified by mass triggered prep LC and all fractions were evaporated to afford the title compound. The compound was analyzed by NMR and HPLC.
Resolution to the (+)- and (-)enantiomers was accomplished by chiral HPLC on a CHIRALCEL™ AD-H column, eluting with 12% isopropanol-hexanes.
Assessment of Biological Properties Compounds of the invention were evaluated for binding to the steroid receptor by a fluorescence polarization competitive binding assay. Detailed descriptions for preparation of recombinant glucocorticoid receptor (GR) complex used in the assay is described in U.S. Patent Application Publication No. US 2003/0017503, filed May 20, 2002, and incorporated herein by reference in its entirety. Preparation of the tetramethylrhodamine (TAMRA)- labeled dexamethasone probe was accomplished using a standard literature procedure (M. Pons et al, J. Steroid Biochem., 1985, 22, pp. 267-273).
A. Glucocorticoid Receptor Competitive Binding Assay Step 1. Characterization of the Fluorescent Probe The wavelengths for maximum excitation and emission of the fluorescent probe should first be measured. An example of such a probe is rhodamine (TAMRA)-labeled dexamethasone.
The affinity of the probe for the steroid receptor was then determined in a titration experiment. The fluorescence polarization value of the probe in assay buffer was measured on an SLM-8100 fluorometer using the excitation and emission maximum values described above. Aliquots of expression vector lysate were added and fluorescence polarization was measured after each addition until no further change in polarization value was observed. Non-linear least squares regression analysis was used to calculate the dissociation constant of the probe from the polarization values obtained for lysate binding to the probe.
Step 2. Screening for Inhibitors of Probe Binding This assay uses fluorescence polarization (FP) to quantitate the ability of test compounds to compete with tetramethylrhodamine (TAMRA)-labeled dexamethasone for binding to a human glucocorticoid receptor (GR) complex prepared from an insect expression system. The assay buffer was: 10 mM TES, 50 mM KCl, 20 mM Na2MoO4 »2H2O, 1.5 mM EDTA, 0.04% w/v CHAPS, 10% v/v glycerol, 1 mM dithiothreitol, pH 7.4. Test compounds were dissolved to 1 mM in neat DMSO and then further diluted to 1 Ox assay concentration in assay buffer supplemented with 10% v/v DMSO. Test compounds were serially diluted at 1Ox assay concentrations in 10% DMSO-containing buffer in 96-well polypropylene plates. Binding reaction mixtures were prepared in 96-well black Dynex microtiter plates by sequential addition of the following assay components to each well: 15 μL of 10x test compound solution, 85 μL of GR-containing baculovirus lysate diluted 1 : 170 in assay buffer, and 50 μL of 15 nM TAMRA-labeled dexamethasone. Positive controls were reaction mixtures containing no test compound; negative controls (blanks) were reaction mixtures containing 0.7 μM to 2 μM dexamethasone. The binding reactions were incubated for 1 hour at room temperature and then read for fluorescence polarization in the LJL Analyst set to 550 nm excitation and 580 nm emission, with the Rhodamine 561 dichroic mirror installed. IC50 values were determined by iterative non-linear curve fitting of the FP signal data to a 4- parameter logistic equation.
Compounds found to bind to the glucocorticoid receptor may be evaluated for binding to the progesterone receptor (PR), estrogen receptor (ER), and mineralocorticoid receptors to evaluate the compound's selectivity for GR. The protocols for PR and MR are identical to the above GR method, with the following exceptions: PR insect cell lysate is diluted 1 :7.1 and MR lysate diluted 1 :9.4. PR probe is TAMRA-labeled mifepristone, used at a final concentration of 5 nM in the assay, and the negative controls (blanks) were reactions containing mifepristone at 0.7 μM to 2 μM.
The ER protocol is similar to the above protocols, but uses PanVera kit receptor, fluorescein- labeled probe. The assay components are made in the same volumes as above, to produce final assay concentrations for ER of 15 nM and ES2 probe of 1 nM. In addition, the component order of addition is modified from the above assays: probe is added to the plate first, followed by receptor and test compound. The plates are read in the LJL Analyst set to 485 nm excitation and 530 nm emission, with the Fluorescein 505 dichroic mirror installed.
Compounds found to bind to the glucocorticoid receptor may be evaluated for dissociation of transactivation and transrepression by assays cited in the Background of the Invention (CM. Bamberger and H.M. Schulte, Eur. J. Clin. Invest., 2000, 30 (suppl. 3) 6-9) or by the assays described below.
B. Glucocorticoid Receptor Cell Assays 1. Induction of Aromatase in Fibroblasts (Cell Assay for Transactivation)
Dexamethasone, a synthetic ligand to the glucocorticoid receptor (GR), induces expression of aromatase in human foreskin fibroblast cells. The activity of aromatase is measured by the conversion of testosterone to estradiol in culture media. Compounds that exhibit binding to GR are evaluated for their ability to induce aromatase activity in human foreskin fibroblasts.
Human foreskin fibroblast cells (ATCC Cat. No. CRL-2429, designation CCDl 12SK) are plated on 96 well plates at 50,000 cells per well 5 days before use, in Iscove's Modified Dulbecco's Media (GibcoBRL Life Technologies Cat No. 12440-053) supplemented with 10% charcoal filtered FBS (Clonetech Cat No. SH30068) and Gentamycin (GibcoBRL Life Technologies Cat. No. 15710-064). On the day of the experiment, the media in the wells is replaced with fresh media. Cells are treated with test compounds to final concentrations of 10"5 M to 10"8 M, and testosterone to a final concentration of 300 ng/mL. Each well has a total volume of 100 μL. Samples are made in duplicates. Control wells include: (a) wells that receive testosterone only, and (b) wells that receive testosterone plus 2 μM of dexamethasone to provide maximum induction of aromatase. Plates are incubated at 37°C overnight (15 to 18 hours), and supernatants are harvested at the end of incubation. Estradiol in the supernatant is measured using ELISA kits for estradiol (made by ALPCO, obtained from American Laboratory Products Cat. No. 020-DR-2693) according to the manufacture's instruction. The amount of estradiol is inversely proportional to the ELISA signals in each well. The extent of aromatase induction by test compounds is expressed as a relative percentage to dexamethasone. EC50 values of test compounds are derived by non- linear curve fitting. 2. Inhibition of IL-6 Production in Fibroblasts (Cell Assay for Transrepression)
Human foreskin fibroblast cells produce IL-6 in response to stimulation by pro-inflammatory cytokine IL-I . This inflammatory response, as measured by the production of IL-6, can be effectively inhibited by dexamethasone, a synthetic ligand to the glucocorticoid receptor (GR). Compounds that exhibit binding to GR are evaluated for their ability to inhibit IL-6 production in human foreskin fibroblasts.
Human foreskin fibroblast cells (ATCC Cat. No. CRL-2429) are plated on 96 well plates at 5,000 cells per well the day before use, in Iscove's Modified Dulbecco's Media (GibcoBRL Life Technologies Cat. No. 12440-053) supplemented with 10% charcoal filtered FBS (Clonetech Cat. No. SH30068) and Gentamycin (GibcoBRL Life Technologies Cat. No. 15710-064). On the next day, media in the wells is replaced with fresh media. Cells are treated with IL-I (rhIL-lα, R&D Systems Cat. No. 200-LA) to a final concentration of 1 ng/mL, and with test compounds to final concentrations of 10~5 M to 10 8 M, in a total volume of 200 μL per well. Samples are done in duplicates. Background control wells do not receive test compounds or IL- 1. Positive control wells receive IL- 1 only and represent maximum (or 100%) amount of IL-6 production. Plates are incubated at 37°C overnight (15 to 18 hours), and supernatants are harvested at the end of incubation. IL-6 levels in the supernatants are determined by the ELISA kits for IL-6 (MedSystems Diagnostics GmbH, Vienna, Austria, Cat. No. BMS213TEN) according to manufacture's instructions. The extent of inhibition of IL-6 by test compounds is expressed in percentage relative to positive controls. IC50 values of test compounds are derived by non-linear curve fitting.
Evaluation of agonist or antagonist activity of compounds binding to the glucocorticoid receptor may be determined by any of the assays.
3. Modulation of Tyrosine Aminotransferase (TAT) Induction in Rat Hepatoma Cells Testing of compounds for agonist or antagonist activity in induction of tyrosine aminotransferase (TAT) in rat hepatoma cells.
H4-II-E-C3 cells were incubated overnight in 96 well plates (20,000 cells/100 μL/well) in MEM medium containing 10% heat inactivated FBS and 1% nonessential amino acids. On the next day, cells were stimulated with the indicated concentrations of dexamethasone or test compound (dissolved in DMSO, final DMSO concentration 0.2%) for 18 hours. Control cells were treated with 0.2% DMSO. After 18 hours, the cells were lysed in a buffer containing 0.1% Triton X-IOO and the TAT activity was measured in a photometric assay using tyrosine and alpha-ketoglutarate as substrates.
For measuring antagonist activity, the hepatoma cells were pre-stimulated by addition of dexamethasone (concentration ranges from 3 x 10"9 M to 3 x 10"8 M) shortly before the test compound was applied to the cells. The steroidal non-selective GR/PR antagonist mifepristone was used as control.
4. Modulation ofMMTV-Luc Induction in HeLa Cells
Testing of compounds for agonist or antagonist activity in stimulation of MMTV-(mouse mammary tumor virus) promoter in HeLa cells.
HeLa cells were stably co-transfected with the pHHLuc-plasmid containing a fragment of the MMTV-LTR (-200 to +100 relative to the transcription start site) cloned in front of the luciferase gene (Norden, 1988) and the pcDNA3.1 plasmid (Invitrogen) constitutively expressing the resistance for the selective antibiotic GENETICIN®. Clones with best induction of the MMTV-promoter were selected and used for further experiments.
Cells were cultured overnight in DMEM medium without phenol red, supplemented with 3% CCS (charcoal treated calf serum) and then transferred to 96 well plates (15,000 cells/100 μL/well). On the next day, activation of the MMTV-promoter was stimulated by addition of test compound or dexamethasone dissolved in DMSO (final concentration 0.2%). Control cells were treated with DMSO only. After 18 hours, the cells were lysed with cell lysis reagent (Promega, Cat. No. E1531), luciferase assay reagent (Promega, Cat. No. E1501) was added and the glow luminescence was measured using a lumino meter (BMG, Offenburg).
For measuring antagonist activity, the MMTV-promoter was pre-stimulated by adding dexamethasone (3 x 109 M to 3 x 10 8 M) shortly before the test compound was applied to the cells. The steroidal non-selective GR/PR antagonist mifepristone was used as control. 5. Modulation of IL-8 Production in U937 Cells
Testing of compounds for agonist or antagonist activity in GR-mediated inhibition of LPS- induced IL-8 secretion in U-937 cells.
U-937 cells were incubated for 2 to 4 days in RPMI1640 medium containing 10% CCS (charcoal treated calf serum). The cells were transferred to 96 well plates (40,000 cells/100 μL/well) and stimulated with 1 μg/mL LPS (dissolved in PBS) in the presence or absence of dexamethasone or test compound (dissolved in DMSO, final concentration 0.2%). Control cells were treated with 0.2% DMSO. After 18 hours, the IL-8 concentration in the cell supernatant was measured by ELISA, using the "OptEIA human IL-8 set" (Pharmingen, Cat. No. 2654KI).
For measuring antagonist activity, the LPS-induced IL-8 secretion was inhibited by adding dexamethasone (3 x 10"9 M to 3 x 10"8 M) shortly before the test compound was applied to the cells. The steroidal non-selective GR/PR antagonist mifepristone was used as control.
6. Modulation oflCAM-Luc Expression in HeLa Cells
Testing of compounds for agonist or antagonist activity in inhibition of TNF-alpha-induced activation of the ICAM-promoter in HeLa cells.
HeLa cells were stably co-transfected with a plasmid containing a 1.3 kb fragment of the human ICAM-promoter (-1353 to -9 relative to the transcription start site, Ledebur and Parks, 1995) cloned in front of the luciferase gene and the pcDNA3.1 plasmid (Invitrogen) which constitutively expresses the resistance for the antibiotic GENETICIN®. Clones with best induction of the ICAM-promoter were selected and used for further experiments. Cells were transferred to 96 well plates (15,000 cells/ 100 μL/well) in DMEM medium supplemented with 3% CCS. On the following day the activation of the ICAM-promoter was induced by addition of 10 ng/mL recombinant TNF-alpha (R&D System, Cat. No. 210-TA). Simultaneously the cells were treated with the test compound or dexamethasone (dissolved in DMSO, final concentration 0.2%). Control cells were treated with DMSO only. After 18 hours, the cells were lysed with cell lysis reagent (Promega, Cat. No. E1531), luciferase assay reagent (Promega, Cat. No. E1501) was added and glow luminescence was measured using a lumino meter (BMG, Offenburg).
For measuring antagonist activity, the TNF-alpha-induced activation of the ICAM-promoter was inhibited by adding dexamethasone (3 x 10~9 M to 3 x 10~8 M) shortly before the test compound was applied to the cells. The steroidal non-selective GR/PR antagonist mifepristone was used as control.
In general, the preferred potency range (IC50) in the above assays is between 0.1 nM and 10 μM, the more preferred potency range is 0.1 nM to 1 μM, and the most preferred potency range is 0.1 nM to 100 nM.
Representative compounds of the invention have been tested and have shown activity as modulators of the glucocorticoid receptor function in one or more of the above assays. For example, the following compounds of the invention have demonstrated potent activity (IC50 = 100 nM or less) in the GR binding assay:
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
N-[I -(3-Cyano-6-methyl-4-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]- 2,4,6-trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(5-nitropyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-ylaminomethyl)ethyl]benzenesulfonamide; 2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-ylaminomethyl)ethyl]benzenesulfonamide
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(2-methylquinolin-8- ylamino)methyl]ethyl}benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
N-{(5)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N- {(5)- 1 -[(lH-Indol-4-ylamino)methyl]propyl} -2,4,6-trimethylbenzenesulfonamide;
N-(OS)- 1 - { [ 1 -(4-Fluorophenyl)- lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide;
2-Amino-4,6-dichloro-N-((5)- 1 - { [ 1 -(4-fluorophenyl)- lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide; and
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide.
In addition, the following compounds of the invention have been tested and have shown activity as an agonist of the glucocorticoid receptor function in one or more of the above assays:
N-{(5)-l-[(lH-Indol-4-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
2-Amino-4,6-dichloro-N-((S)- 1 - { [ 1 -(4-fluorophenyl)- lH-indazol-5- ylamino]methyl}propyl)benzenesulfonamide; and
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide.
7. Inhibition of Osteocalcin Production from Osteoblast Cell Line MG-63 Human osteosarcoma MG-63 cells (ATCC, Cat. No. CRL-1427) are plated on 96 well plates at 20,000 cells per well the day before use in 200 μL media of 99% D-MEM/F-12 (Gibco- Invitrogen, Cat. No. 11039-021), supplemented with 1% penicillin and streptomycin (Gibco- Invitrogen, Cat. No. 15140-122), 10 μg/mL Vitamin C (Sigma, Cat. No. A-4544), and 1% charcoal filtered Fetal Bovine Serum (HyClone, Cat. No. SH30068.02). The next day, wells are replaced with fresh media. Cells are treated with Vitamin D (Sigma, Cat. No. D 1530) to a final concentration of 10 nM, and with the test compounds in concentrations of 10 6 M to 10 9 M, in a total volume of 200 μL per well. Samples are done in duplicates. Background control wells do not receive Vitamin D or compounds. Positive control wells receive Vitamin D only, without compounds, and represent maximum (100%) amount of osteocalcin production. Plates are incubated at 37°C incubator for 48 hours and supernatants are harvested at the end of incubation. Amounts of osteocalcin in the supernatants are determined by the Glype osteocalcin ELISA kit (Zymed, Cat. No. 99-0054) according to manufacture's protocol. Inhibition of osteocalcin by test compounds is expressed in percentage relative to positive controls. IC50 values of the test compounds are derived by non- lineal curve fitting.
The invention also provides methods of modulating the glucocorticoid receptor function in a patient comprising administering to the patient a compound according to the invention. If the purpose of modulating the glucocorticoid receptor function in a patient is to treat a disease- state or condition, the administration preferably comprises a therapeutically or pharmaceutically effective amount of a pharmaceutically acceptable compound according to the invention. If the purpose of modulating the glucocorticoid receptor function in a patient is for a diagnostic or other purpose (e.g., to determine the patient's suitability for therapy or sensitivity to various sub-therapeutic doses of the compounds according to the invention), the administration preferably comprises an effective amount of a compound according to the invention, that is, the amount necessary to obtain the desired effect or degree of modulation.
Methods of Therapeutic Use
As pointed out above, the compounds of the invention are useful in modulating the glucocorticoid receptor function. In doing so, these compounds have therapeutic use in treating disease-states and conditions mediated by the glucocorticoid receptor function or that would benefit from modulation of the glucocorticoid receptor function. As the compounds of the invention modulate the glucocorticoid receptor function, they have very useful anti-inflammatory and antiallergic, immune-suppressive, and anti-proliferative activity and they can be used in patients as drugs, particularly in the form of pharmaceutical compositions as set forth below, for the treatment of disease-states and conditions.
The agonist compounds according to the invention can be used in patients as drugs for the treatment of the following disease-states or indications that are accompanied by inflammatory, allergic, and/or proliferative processes: (i) Lung diseases: chronic, obstructive lung diseases of any genesis, particularly bronchial asthma and chronic obstructive pulmonary disease (COPD); adult respiratory distress syndrome (ARDS); bronchiectasis; bronchitis of various genesis; all forms of restrictive lung diseases, particularly allergic alveolitis; all forms of lung edema, particularly toxic lung edema; all forms of interstitial lung diseases of any genesis, e.g., radiation pneumonitis; and sarcoidosis and granulomatoses, particularly
Boeck disease;
(ii) Rheumatic diseases or autoimmune diseases or joint diseases: all forms of rheumatic diseases, especially rheumatoid arthritis, acute rheumatic fever, and polymyalgia rheumatica; reactive arthritis; rheumatic soft tissue diseases; inflammatory soft tissue diseases of other genesis; arthritic symptoms in degenerative joint diseases
(arthroses); traumatic arthritis; collagenoses of any genesis, e.g., systemic lupus erythematosus, scleroderma, polymyositis, dermatomyositis, Sjogren syndrome, Still disease, and Felty syndrome;
(iii) Allergic diseases: all forms of allergic reactions, e.g., angioneurotic edema, hay fever, insect bites, allergic reactions to drugs, blood derivatives, contrast agents, etc., anaphylactic shock (anaphylaxis), urticaria, angioneurotic edema, and contact dermatitis;
(iv) Vasculitis diseases: panarteritis nodosa, polyarteritis nodosa, arteritis temporalis,
Wegner granulomatosis, giant cell arthritis, and erythema nodosum; (v) Dermatological diseases: atopic dermatitis, particularly in children; psoriasis; pityriasis rubra pilaris; erythematous diseases triggered by various noxa, e.g., rays, chemicals, burns, etc.; bullous dermatoses; diseases of the lichenoid complex; pruritus (e.g., of allergic genesis); seborrheic dermatitis; rosacea; pemphigus vulgaris; erythema multiforme exudativum; balanitis; vulvitis; hair loss, such as occurs in alopecia areata; and cutaneous T cell lymphomas;
(vi) Renal diseases: nephrotic syndrome; and all types of nephritis, e.g., glomerulonephritis;
(vii) Hepatic diseases: acute liver cell disintegration; acute hepatitis of various genesis, e.g., viral, toxic, drug-induced; and chronically aggressive and/or chronically intermittent hepatitis;
(viii) Gastrointestinal diseases: inflammatory bowel diseases, e.g., regional enteritis (Crohn disease), colitis ulcerosa; gastritis; peptic esophagitis (refluxoesophagitis); and gastroenteritis of other genesis, e.g., nontropical sprue;
(ix) Proctological diseases: anal eczema; fissures; hemorrhoids; and idiopathic proctitis; (x) Eye diseases: allergic keratitis, uveitis, or iritis; conjunctivitis; blepharitis; neuritis nervi optici; choroiditis; and sympathetic ophthalmia; (xi) Diseases of the ear, nose, and throat (ENT) area: allergic rhinitis or hay fever; otitis externa, e.g., caused by contact eczema, infection, etc.; and otitis media; (xii) Neurological diseases: brain edema, particularly tumor-related brain edema; multiple sclerosis; acute encephalomyelitis; meningitis; acute spinal cord injury; stroke; and various forms of seizures, e.g., nodding spasms; (xiϋ) Blood diseases: acquired hemolytic anemia; and idiopathic thrombocytopenia;
(xiv) Tumor diseases: acute lymphatic leukemia; malignant lymphoma; lymphogranulomatoses; lymphosarcoma; extensive metastases, particularly in mammary, bronchial, and prostatic carcinoma;
(xv) Endocrine diseases: endocrine ophthalmopathy; endocrine orbitopathia; thyrotoxic crisis; Thyroiditis de Quervain; Hashimoto thyroiditis; Morbus Basedow; granulomatous thyroiditis; struma lymphomatosa; and Grave disease; (xvi) Organ and tissue transplantations and graft- versus-host diseases; (xvii) Severe states of shock, e.g., septic shock, anaphylactic shock, and systemic inflammatory response syndrome (SIRS); (xviii) Substitution therapy in: congenital primary adrenal insufficiency, e.g., adrenogenital syndrome; acquired primary adrenal insufficiency, e.g., Addison disease, autoimmune adrenalitis, post-infection, tumors, metastases, etc.; congenital secondary adrenal insufficiency, e.g., congenital hypopituitarism; and acquired secondary adrenal insufficiency, e.g., post-infection, tumors, metastases, etc.;
(xix) Pain of inflammatory genesis, e.g., lumbago; and
(xx) various other disease-states or conditions including type I diabetes (insulin-dependent diabetes), osteoarthritis, Guillain-Barre syndrome, restenosis following percutaneous transluminal coronary angioplasty, Alzheimer disease, acute and chronic pain, atherosclerosis, reperfusion injury, bone resorption diseases, congestive heart failure, myocardial infarction, thermal injury, multiple organ injury secondary to trauma, acute purulent meningitis, necrotizing enterocolitis and syndromes associated with hemodialysis, leukopheresis, and granulocyte transfusion.
In addition, the compounds according to the invention can be used for the treatment of any other disease-states or conditions not mentioned above which have been treated, are treated, or will be treated with synthetic glucocorticoids (see, e.g., H.J. Hatz, Glucocorticoide: Immunologische Grundlagen, Pharmakologie und Therapierichtlinien [Glucocorticoids: Immunological Fundamentals, Pharmacology, and Therapeutic Guidelines], Stuttgart: Verlagsgesellschaft mbH, 1998, which is hereby incorporated by reference in its entirety). Most or all of the indications (i) through (xx) mentioned above are described in detail in H.J. Hatz, Glucocorticoide: Immunologische Grundlagen, Pharmakologie und Therapierichtlinien. Furthermore, the compounds of the invention can also be used to treat disorders other than those listed above or mentioned or discussed herein, including in the Background of the Invention.
The antagonist compounds according to the invention, whether full antagonists or partial antagonists, can be used in patients as drugs for the treatment of the following disease-states or indications, without limitation: type II diabetes (non-insulin-dependent diabetes); obesity; cardiovascular diseases; hypertension; arteriosclerosis; neurological diseases, such as psychosis and depression; adrenal and pituitary tumors; glaucoma; and Cushing syndrome based on an ACTH-secreting tumor like pituitary adenoma. In particular, the compounds of the invention are useful for treating obesity and all disease-states and indications related to a deregulated fatty acids metabolism such as hypertension, atherosclerosis, and other cardiovascular diseases. Using the compounds of the invention that are GR antagonists, it should be possible to antagonize both the carbohydrate metabolism and fatty acids metabolism. Thus, the antagonist compounds of the invention are useful in treating all disease-states and conditions that involve increased carbohydrate, protein, and lipid metabolism and would include disease-states and conditions leading to catabolism like muscle frailty (as an example of protein metabolism).
Methods of Diagnostic Use
The compounds of the invention may also be used in diagnostic applications and for commercial and other purposes as standards in competitive binding assays. In such uses, the compounds of the invention may be used in the form of the compounds themselves or they may be modified by attaching a radioisotope, luminescence, fluorescent label or the like in order to obtain a radioisotope, luminescence, or fluorescent probe, as would be known by one of skill in the art and as outlined in Handbook of Fluorescent Probes and Research Chemicals, 6th Edition, R.P. Haugland (ed.), Eugene: Molecular Probes, 1996; Fluorescence and Luminescence Probes for Biological Activity, W.T. Mason (ed.), San Diego: Academic Press, 1993; Receptor-Ligand Interaction, A Practical Approach, E. C. Hulme (ed.), Oxford: IRL Press, 1992, each of which is hereby incorporated by reference in their entireties.
General Administration and Pharmaceutical Compositions When used as pharmaceuticals, the compounds of the invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared using procedures well known in the pharmaceutical art and comprise at least one compound of the invention. The compounds of the invention may also be administered alone or in combination with adjuvants that enhance stability of the compounds of the invention, facilitate administration of pharmaceutical compositions containing them in certain embodiments, provide increased dissolution or dispersion, increased inhibitory activity, provide adjunct therapy, and the like. The compounds according to the invention may be used on their own or in conjunction with other active substances according to the invention, optionally also in conjunction with other pharmacologically active substances. In general, the compounds of this invention are administered in a therapeutically or pharmaceutically effective amount, but may be administered in lower amounts for diagnostic or other purposes. In particular, the compounds of the invention are useful in combination with glucocorticoids or corticosteroids. As pointed out above, standard therapy for a variety of immune and inflammatory disorders includes administration of corticosteroids, which have the ability to suppress immunologic and inflammatory responses. (A.P. Truhan et al, Annals of Allergy, 1989, 62, pp. 375-391; J.D. Baxter, Hospital Practice, 1992, 27, pp. 111-134; R.P. Kimberly, Curr. Opin. Rheumatol., 1992, 4, pp. 325-331; M.H. Weisman, Curr. Opin. Rheumatol., 1995, 7, pp. 183-190; W. Sterry, Arch. Dermatol. Res., 1992, 284 (Suppl.), pp. S27-S29). While therapeutically beneficial, however, the use of corticosteroids is associated with a number of side effects, ranging from mild to possibly life threatening, especially with prolonged and/or high dose steroid usage. Accordingly, methods and compositions that enable the use of a lower effective dosage of corticosteroids (referred to as the "steroid sparing effect") would be highly desirable to avoid unwanted side effects. The compounds of the invention provide such a steroid sparing effect by achieving the desired therapeutic effect while allowing the use of lower doses and less frequent administration of glucocorticoids or corticosteroids.
Administration of the compounds of the invention, in pure form or in an appropriate pharmaceutical composition, can be carried out using any of the accepted modes of administration of pharmaceutical compositions. Thus, administration can be, for example, orally, buccally (e.g., sublingually), nasally, parenterally, topically, transdermally, vaginally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as, for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages. The pharmaceutical compositions will generally include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include other medicinal agents, pharmaceutical agents, carriers, adjuvants, diluents, vehicles, or combinations thereof. Such pharmaceutically acceptable excipients, carriers, or additives as well as methods of making pharmaceutical compositions for various modes or administration are well-known to those of skill in the art. The state of the art is evidenced, e.g., by Remington: The Science and Practice of Pharmacy. 20th Edition, A. Gennaro (ed.), Lippincott Williams & Wilkins, 2000; Handbook of Pharmaceutical Additives, Michael & Irene Ash (eds.), Gower, 1995; Handbook of Pharmaceutical Excipients. A.H. Kibbe (ed.), American Pharmaceutical Ass'n, 2000; H. C. Ansel and N. G. Popovish, Pharmaceutical Dosage Forms and Drug Delivery Systems, 5th ed., Lea and Febiger, 1990; each of which is incorporated herein by reference in their entireties to better describe the state of the art.
As one of skill in the art would expect, the forms of the compounds of the invention utilized in a particular pharmaceutical formulation will be selected (e.g., salts) that possess suitable physical characteristics (e.g., water solubility) that is required for the formulation to be efficacious.
Pharmaceutical compositions suitable for buccal (sub-lingual) administration include lozenges comprising a compound of the present invention in a flavored base, usually sucrose, and acacia or tragacanth, and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
Pharmaceutical compositions suitable for parenteral administration comprise sterile aqueous preparations of a compound of the present invention. These preparations are preferably administered intravenously, although administration can also be effected by means of subcutaneous, intramuscular, or intradermal injection. Injectable pharmaceutical formulations are commonly based upon injectable sterile saline, phosphate-buffered saline, oleaginous suspensions, or other injectable carriers known in the art and are generally rendered sterile and isotonic with the blood. The injectable pharmaceutical formulations may therefore be provided as a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, including 1,3-butanediol, water, Ringer's solution, isotonic sodium chloride solution, fixed oils such as synthetic mono- or diglycerides, fatty acids such as oleic acid, and the like. Such injectable pharmaceutical formulations are formulated according to the known art using suitable dispersing or setting agents and suspending agents. Injectable compositions will generally contain from 0.1 to 5% w/w of a compound of the invention.
Solid dosage forms for oral administration of the compounds include capsules, tablets, pills, powders, and granules. For such oral administration, a pharmaceutically acceptable composition containing a compound(s) of the invention is formed by the incorporation of any of the normally employed excipients, such as, for example, pharmaceutical grades of mannitol, lactose, starch, pregelatinized starch, magnesium stearate, sodium saccharine, talcum, cellulose ether derivatives, glucose, gelatin, sucrose, citrate, propyl gallate, and the like. Such solid pharmaceutical formulations may include formulations, as are well known in the art, to provide prolonged or sustained delivery of the drug to the gastrointestinal tract by any number of mechanisms, which include, but are not limited to, pH sensitive release from the dosage form based on the changing pH of the small intestine, slow erosion of a tablet or capsule, retention in the stomach based on the physical properties of the formulation, bioadhesion of the dosage form to the mucosal lining of the intestinal tract, or enzymatic release of the active drug from the dosage form.
Liquid dosage forms for oral administration of the compounds include emulsions, microemulsions, solutions, suspensions, syrups, and elixirs, optionally containing pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like. These compositions can also contain additional adjuvants such as wetting, emulsifying, suspending, sweetening, flavoring, and perfuming agents.
Topical dosage forms of the compounds include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants, eye ointments, eye or ear drops, impregnated dressings and aerosols, and may contain appropriate conventional additives such as preservatives, solvents to assist drug penetration and emollients in ointments and creams. Topical application may be once or more than once per day depending upon the usual medical considerations. Furthermore, preferred compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles. The formulations may also contain compatible conventional carriers, such as cream or ointment bases and ethanol or oleyl alcohol for lotions. Such carriers may be present as from about 1% up to about 98% of the formulation, more usually they will form up to about 80% of the formulation.
Transdermal administration is also possible. Pharmaceutical compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. Such patches suitably contain a compound of the invention in an optionally buffered, aqueous solution, dissolved and/or dispersed in an adhesive, or dispersed in a polymer. A suitable concentration of the active compound is about 1% to 35%, preferably about 3% to 15%.
For administration by inhalation, the compounds of the invention are conveniently delivered in the form of an aerosol spray from a pump spray device not requiring a propellant gas or from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, heptafluoropropane, carbon dioxide, or other suitable gas. In any case, the aerosol spray dosage unit may be determined by providing a valve to deliver a metered amount so that the resulting metered dose inhaler (MDI) is used to administer the compounds of the invention in a reproducible and controlled way. Such inhaler, nebulizer, or atomizer devices are known in the art, for example, in PCT International Publication Nos. WO 97/12687 (particularly Figure 6 thereof, which is the basis for the commercial RESPIMAT® nebulizer); WO 94/07607; WO 97/12683; and WO 97/20590, to which reference is hereby made and each of which is incorporated herein by reference in their entireties.
Rectal administration can be effected utilizing unit dose suppositories in which the compound is admixed with low-melting water-soluble or insoluble solids such as fats, cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights, or fatty acid esters of polyethylene glycols, or the like. The active compound is usually a minor component, often from about 0.05 to 10% by weight, with the remainder being the base component.
In all of the above pharmaceutical compositions, the compounds of the invention are formulated with an acceptable carrier or excipient. The carriers or excipients used must, of course, be acceptable in the sense of being compatible with the other ingredients of the composition and must not be deleterious to the patient. The carrier or excipient can be a solid or a liquid, or both, and is preferably formulated with the compound of the invention as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compound. Such carriers or excipients include inert fillers or diluents, binders, lubricants, disintegrating agents, solution retardants, resorption accelerators, absorption agents, and coloring agents. Suitable binders include starch, gelatin, natural sugars such as glucose or β-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
Generally, a therapeutically effective daily dose is from about 0.001 mg to about 15 mg/kg of body weight per day of a compound of the invention; preferably, from about 0.1 mg to about
10 mg/kg of body weight per day; and most preferably, from about 0.1 mg to about 1.5 mg/kg of body weight per day. For example, for administration to a 70 kg person, the dosage range would be from about 0.07 mg to about 1050 mg per day of a compound of the invention, preferably from about 7.0 mg to about 700 mg per day, and most preferably from about 7.0 mg to about 105 mg per day. Some degree of routine dose optimization may be required to determine an optimal dosing level and pattern.
Pharmaceutically acceptable carriers and excipients encompass all the foregoing additives and the like.
Examples of Pharmaceutical Formulations
Figure imgf000090_0001
The finely ground active substance, lactose, and some of the corn starch are mixed together. The mixture is screened, then moistened with a solution of polyvinylpyrrolidone in water, kneaded, wet-granulated and dried. The granules, the remaining corn starch and the magnesium stearate are screened and mixed together. The mixture is compressed to produce tablets of suitable shape and size.
Figure imgf000091_0001
The finely ground active substance, some of the corn starch, lactose, microcrystalline cellulose, and polyvinylpyrrolidone are mixed together, the mixture is screened and worked with the remaining corn starch and water to form a granulate which is dried and screened. The sodium-carboxymethyl starch and the magnesium stearate are added and mixed in and the mixture is compressed to form tablets of a suitable size.
Figure imgf000091_0002
The active substance, corn starch, lactose, and polyvinylpyrrolidone are thoroughly mixed and moistened with water. The moist mass is pushed through a screen with a 1 mm mesh size, dried at about 45°C and the granules are then passed through the same screen. After the magnesium stearate has been mixed in, convex tablet cores with a diameter of 6 mm are compressed in a tablet-making machine. The tablet cores thus produced are coated in known manner with a covering consisting essentially of sugar and talc. The finished coated tablets are polished with wax.
Figure imgf000092_0001
The substance and corn starch are mixed and moistened with water. The moist mass is screened and dried. The dry granules are screened and mixed with magnesium stearate. The finished mixture is packed into size 1 hard gelatine capsules.
Figure imgf000092_0002
The active substance is dissolved in water at its own pH or optionally at pH 5.5 to 6.5 and sodium chloride is added to make it isotonic. The solution obtained is filtered free from pyrogens and the filtrate is transferred under aseptic conditions into ampoules which are then sterilized and sealed by fusion. The ampoules contain 5 mg, 25 mg, and 50 mg of active substance.
Figure imgf000093_0001
The hard fat is melted. At 400C, the ground active substance is homogeneously dispersed therein. The mixture is cooled to 38°C and poured into slightly chilled suppository molds.
Figure imgf000093_0002
The suspension is transferred into a conventional aerosol container with a metering valve. Preferably, 50 μL of suspension are delivered per spray. The active substance may also be metered in higher doses if desired (e.g., 0.02% by weight).
Figure imgf000093_0003
Figure imgf000093_0004
Figure imgf000094_0001
Figure imgf000094_0002
In Examples H, I, J, and K, the powder for inhalation is produced in the usual way by mixing the individual ingredients together.

Claims

We Claim:
1. A compound of Formula (IA)
Figure imgf000095_0001
wherein:
R1 is hydrogen or Ci-C3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo;
R2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently Ci-C5 alkyl, C2-C3 alkenyl, C2- C3 alkynyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C1-Cs alkoxycarbonylamino, C1-Cs alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1- C5 alkyl; or ureido wherein either nitrogen atom is optionally independently substituted with C1-Cs alkyl; or C1-Cs alkylthio,
wherein each substituent group of R is optionally independently substituted with C1- C3 alkyl, halogen, hydroxyl, or amino,
wherein R2 cannot be /?-methylphenyl;
R3 is a hydrogen or C1-Cs alkyl, optionally independently substituted with one, two, or three substituent groups, wherein each substituent group of R3 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R4 and R5 are each independently hydrogen, Ci-C5 alkyl, or phenyl, or R4 and R5 together with the carbon atom they are commonly attached to form a C3-C8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R4 and R5 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R6 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R6 is independently C1-C3 alkyl, C2-C5 alkenyl, C2- C5 alkynyl, heterocyclyl, aryl, heteroaryl, Ci-C5 alkoxy, acyl, acylamino, aminocarbonyl, C1-C3 alkylamioncarbonyl, C1-C3 dialkylaminocarbonyl halogen, hydroxy, carboxy, cyano, oxo, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, nitro, or amino wherein the nitrogen atom is optionally independently mono- or di- substituted by Ci-C5 alkyl; or Ci-C5 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R6 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, halogen, hydroxy, oxo, cyano, phenyl, amino, or trifluoromethyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, phenyl optionally substituted with Ci-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
2. The compound of Formula (IA) according to claim 1, wherein:
R1 is hydrogen;
R2 is phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-C3 alkyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by Ci-C5 alkyl, or Ci-C5 alkylthio,
wherein R2 cannot be />-methylphenyl;
R3 is hydrogen;
R4 and R5 are each hydrogen or Ci-C5 alkyl;
R6 is an indolyl, dihydroisoindolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, dihydroisobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, dihydrobenzodioxepinyl, acridinyl, pyrimidinyl or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups, wherein each substituent group of R6 is independently C1-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C1-C3 alkoxy, acylamino, aminocarbonyl, C1-C3 alkylaminocarbonyl, C1-C3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or C1-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, phenyl, or trifluoromethyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, phenyl, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
3. The compound of Formula (IA) according to claim 1, wherein:
R1 is hydrogen;
R2 is a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-Cs alkyl, C1-C3 alkoxy, hydroxy, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-C3 alkyl, or Ci-C3 alkylthio, wherein R2 cannot be p-methylphenyl;
R3 is hydrogen;
R4 and R5 are each hydrogen;
R6 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, dihydrobenzodioxinyl, benzopyranyl, benzothienyl, benzothiazolyl, benzothiophenyl, benzimidazolyl, dihydrobenzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R6 is independently C1-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C1-C3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, oxo, trifluoromethyl, or Ci-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R6 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7; and
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, phenyl, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
4. The compound of Formula (IA) according to claim 1, wherein the compound is selected from:
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-8-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-7-yloxymethyl)ethyl];
N-[l-(Acridin-4-yloxymethyl)-2,2,2-trifluoroethyl]-2,4,6-trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-oxo-2,3-dihydrobenzofuran-6- yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-7-yloxymethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-8-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-methylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-methyl-2-oxo-2H- 1 -benzopyran-7- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-furan-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
N-[l-(3-Cyano-6-methyl-4-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]- 2,4,6-trimethylbenzenesulfonamide; N- { 1 -[4-(4-Chlorophenyl)pyrimidin-2-ylsulfanylmethyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide ;
N-[l-(3-Chloro-5-trifluoromethylpyridin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide ;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(5-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(4-trifluoromethylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[4-(4-methoxyphenyl)pyrimidin-2- ylsulfanylmethyl] ethyl } benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(4-phenoxypyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(5-nitropyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-4-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(pyridin-2-ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro- 1 -(pyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(3-trifluoromethylpyridin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide; 2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-5-ylaminomethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-6-ylaminomethyl)ethyl]benzenesulfonamide;
N- { 1 -[(2,3-Dihydro- 1 ,4-benzodioxin-6-ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide ;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(isoquinolin-5- ylaminomethyl)ethyl]benzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(2-methylquinolin-8- ylamino)methyl]ethyl}benzenesulfonamide;
N-[l-(Benzothiazol-7-ylaminomethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- l-[(2-methyl-l,3-dioxo-2,3-dihydro-lH-isoindol-5- ylamino)methyl]ethyl}benzenesulfonamide;
N-{l-[(l,l-Dioxo-lH-lλ6-benzo[&]thiophen-5-ylamino)methyl]-2,2,2-trifluoroethyl}-2,4,6- trimethylbenzenesulfonamide;
N- { 1 -[(3,4-Dihydro-2H- 1 ,5-benzodioxepin-7-ylamino)methyl]-2,2,2-trifluoroethyl} -2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(methylpyridin-2- ylamino)methyl]ethyl}benzenesulfonamide;
2,4,6-Trimethyl-N-(2,2,2-trifluoro- 1 - { [(4-methoxybenzyl)pyridin-2- ylamino]methyl}ethyl)benzenesulfonamide; N-(l-{[Benzyl-(4-methylpyridin-2-yl)-amino]methyl}-2,2,2-trifluoroethyl)-2,4,6- trimethylbenzenesulfonamide ;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- 1 -[(methylpyridin-4- ylamino)methyl]ethyl}benzenesulfonamide;
2,4,6-Trimethyl-N-(2,2,2-trifluoro-l-{[(6-methoxypyridin-2- yl)methylamino]methyl}ethyl)benzenesulfonamide;
N-[l-(4,6-Dimethylpyrimidin-2-ylsulfanylmethyl)-2,2,2-trifluoroethyl]-2,4,6- trimethylbenzenesulfonamide;
2,4,6-Trimethyl-N- {2,2,2-trifluoro- l-[(2-oxo-2,3-dihydro-lH-benzimidazol-5- ylamino)methyl]ethyl}benzenesulfonamide;
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(4-thiophen-2-ylpyrimidin-2- ylsulfanylmethyl)ethyl]benzenesulfonamide; and
2,4,6-Trimethyl-N-[2,2,2-trifluoro-l-(quinolin-4-yloxymethyl)ethyl]benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
5. A compound of Formula (IB)
Figure imgf000103_0001
wherein:
R1 is hydrogen or C1-C3 alkyl, each optionally independently substituted with one, two, or three substituent groups selected from hydroxy, halogen, or oxo; R2 is aryl optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently Ci-C5 alkyl, C2-C3 alkenyl, C2- C3 alkynyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, acylamino, C1-Cs alkoxycarbonylamino, C1-Cs alkylsulfonylamino, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1- C5 alkyl; or ureido wherein either nitrogen atom is optionally independently substituted with Ci-C5 alkyl; or Ci-C5 alkylthio,
wherein each substituent group of R2 is optionally independently substituted with C1- C3 alkyl, halogen, hydroxyl, or amino,
wherein R2 cannot be />-methylphenyl;
R3 is Ci-Cs alkyl independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R3 is independently C3-C6 cycloalkyl, aryl, halogen, trifluoromethyl, trifluoromethoxy, or trifluoromethylthio,
wherein R cannot be a trifluoromethyl;
R4 is a hydrogen or Ci-C5 alkyl, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R4 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
wherein R4 cannot be a trifluoromethyl; R5 and R6 are each independently hydrogen, C1-Cs alkyl or phenyl or R5 and R6 together with the carbon atom they are commonly attached to form a C3-C8 spiro cycloalkyl ring, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R5 and R6 is independently selected from halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and
R8 is a heteroaryl group optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently C1-C3 alkyl, C2-C5 alkenyl, C2- C5 alkynyl, heterocyclyl, aryl, heteroaryl, C1-Cs alkoxy, acyl, acylamino, aminocarbonyl, Ci-C3 alkylamioncarbonyl, Ci-C3 dialkylaminocarbonyl halogen, hydroxy, carboxy, cyano, trifluoromethyl, trifluoromethoxy, trifluoromethylthio, nitro, or amino wherein the nitrogen atom is optionally independently mono- or di- substituted by Ci-C5 alkyl; or Ci-C5 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R8 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, Ci-C3 alkoxy, halogen, hydroxy, oxo, cyano, amino, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
6. The compound of Formula (IB) according to claim 5, wherein:
R1 is hydrogen;
R > 2 i •s phenyl, or naphthyl group, each optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-C3 alkyl, C1-Cs alkoxy, hydroxy, nitro, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-Cs alkyl, or Ci-C5 alkylthio,
wherein R2 cannot be /?-methylphenyl;
R3 is Ci-C5 alkyl independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R3 is independently C3-C8 cycloalkyl, halogen, trifluoromethyl, or trifluoromethoxy,
wherein R3 cannot be a trifluoromethyl;
R4 is hydrogen;
R5 and R6 are each hydrogen or Ci-C5 alkyl;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, Ci-C5 alkyl, or phenyl; wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from C1-C3 alkyl, C1-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl; and
R8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently C1-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, C1-C3 alkoxy, acylamino, aminocarbonyl, C1-C3 alkylaminocarbonyl, C1-C3 dialkylaminocarbonyl, fluoro, chloro, bromo, cyano, trifluoromethyl, or C1-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
7. The compound of Formula (IB) according to claim 5, wherein:
R1 is hydrogen;
R > 2 i •s a phenyl group optionally independently substituted with one, two, three, four, or five substituent groups,
wherein each substituent group of R2 is independently C1-Cs alkyl, C1-C3 alkoxy, hydroxy, trifluoromethyl, trifluoromethoxy, halogen, cyano, or amino wherein the nitrogen atom is optionally independently mono- or di-substituted by C1-C3 alkyl, or Ci-C3 alkylthio; wherein R2 can not be />-methylphenyl;
R3 is methyl, ethyl, isopropyl, or tert-butyl;
R4 is hydrogen;
R5 and R6 are each hydrogen;
X is O, S wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone, or NR7;
R7 is H, Ci-C5 alkyl, or phenyl,
wherein each substituent group of R7 is optionally independently substituted with one, two, or three substituent groups selected from Ci-C3 alkyl, Ci-C3 alkoxy, hydroxy, oxo, cyano, amino, or trifluoromethyl;
R8 is an indolyl, azaindolyl, diazaindolyl, imidazolyl, dihydrobenzofuranyl, benzofuranyl, benzothienyl, benzimidazolyl, isoquinolinyl, quinolinyl, tetrahydroquinolinyl, tetrahydroquinoxalinyl, or pyridinyl group, each optionally independently substituted with one, two, or three substituent groups,
wherein each substituent group of R8 is independently Ci-C3 alkyl, morpholinyl, piperdinyl, phenyl, pyridinyl, pyrimidinyl, Ci-C3 alkoxy, acylamino, fluoro, chloro, bromo, cyano, trifluoromethyl, or Ci-C3 alkylthio wherein the sulfur atom is optionally oxidized to a sulfoxide or sulfone,
wherein each substituent group of R8 is optionally independently substituted with a substituent group selected from methyl, methoxy, fluoro, chloro, bromo, oxo, or trifluoromethyl,
or a tautomer, prodrug, solvate, or salt thereof.
8. The compound of Formula (IB) according to claim 5, wherein the compound is selected from:
N-{(5)-l-[(lH-Indol-7-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-4-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-6-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-{(5)-l-[(lH-Indol-5-ylamino)methyl]propyl}-2,4,6-trimethylbenzenesulfonamide;
N-((5)-l-{[l-(4-Fluorophenyl)-lH-indazol-5-ylamino]methyl}propyl)-2,4,6- trimethylbenzenesulfonamide;
N- {(5)- 1 -[(lH-Indazol-5-ylamino)methyl]propyl} -2,4,6-trimethylbenzenesulfonamide;
N- [(S)- 1 -[(lH-Indazol-6-ylamino)methyl]propyl} -2,4,6-trimethylbenzenesulfonamide;
2-Amino-4,6-dichloro-N-((5)- 1 - {[ 1 -(4-fluorophenyl)- lH-indazol-5-ylamino]methyl}propyl)- benzenesulfonamide;
2-Amino-4,6-dichloro-N-{(5)-l-[(lH-indol-4-ylamino)methyl]propyl}benzenesulfonamide; and
2-Amino-4,6-dichloro-N- [(S)- 1 -[( 1 -oxo- 1 ,3-dihydroisobenzofuran-5- ylamino)methyl]propyl}benzenesulfonamide,
or a tautomer, prodrug, solvate, or salt thereof.
PCT/US2008/059528 2007-04-10 2008-04-07 Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof WO2008124665A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2010503136A JP2010523689A (en) 2007-04-10 2008-04-07 Glucocorticoid mimetics, process for producing the same, pharmaceutical composition and use thereof
CA002683647A CA2683647A1 (en) 2007-04-10 2008-04-07 Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US12/520,997 US20100063282A1 (en) 2007-04-10 2008-04-07 Glucocorticoid Mimetics, Methods of Making Them, Pharmaceutical Compositions, and Uses Thereof
EP08745203A EP2136634A4 (en) 2007-04-10 2008-04-07 Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US92271807P 2007-04-10 2007-04-10
US60/922,718 2007-04-10

Publications (1)

Publication Number Publication Date
WO2008124665A1 true WO2008124665A1 (en) 2008-10-16

Family

ID=39831380

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/059528 WO2008124665A1 (en) 2007-04-10 2008-04-07 Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof

Country Status (5)

Country Link
US (1) US20100063282A1 (en)
EP (1) EP2136634A4 (en)
JP (1) JP2010523689A (en)
CA (1) CA2683647A1 (en)
WO (1) WO2008124665A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7728030B2 (en) 2006-12-21 2010-06-01 Astrazeneca Ab Chemical compounds 572
US20100144713A1 (en) * 2008-01-17 2010-06-10 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
US8030340B2 (en) 2006-11-23 2011-10-04 Astrazeneca Ab Indazolyl sulphonamide derivatives useful as glucocorticoid modulators
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
US8211930B2 (en) 2008-05-20 2012-07-03 Astrazeneca Ab Phenyl and benzodioxinyl substituted indazoles derivatives
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
US8921392B2 (en) 2007-01-10 2014-12-30 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US9096569B2 (en) 2008-07-25 2015-08-04 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US9415043B2 (en) 2013-03-15 2016-08-16 Aerie Pharmaceuticals, Inc. Combination therapy
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US9951059B2 (en) 2009-05-01 2018-04-24 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US10550087B2 (en) 2015-11-17 2020-02-04 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US10624882B2 (en) 2006-09-20 2020-04-21 Aerie Pharmaceuticals, Inc. Rho kinase inhibitors
US10858339B2 (en) 2017-03-31 2020-12-08 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US11389441B2 (en) 2016-08-31 2022-07-19 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
US11427563B2 (en) 2018-09-14 2022-08-30 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050203128A1 (en) * 2004-03-13 2005-09-15 Boehringer Ingelheim Pharmaceuticals, Inc. Glucocorticoid mimetics, methods of making them, pharmaceutical compositions and uses thereof
US20060205712A1 (en) * 2002-06-07 2006-09-14 Federico Calvani Basic non-peptide bradykinin antagonists and pharmaceutical compositions therefrom

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005300150A1 (en) * 2004-10-29 2006-05-04 Astrazeneca Ab Novel sulphonamide derivatives as glucocorticoid receptor modulators for the treatment of inflammatory diseases
JP2008525525A (en) * 2004-12-27 2008-07-17 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Glucocorticoid mimetic, its production, pharmaceutical composition and use
TW200815361A (en) * 2005-10-20 2008-04-01 Astrazeneca Ab Chemical compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060205712A1 (en) * 2002-06-07 2006-09-14 Federico Calvani Basic non-peptide bradykinin antagonists and pharmaceutical compositions therefrom
US20050203128A1 (en) * 2004-03-13 2005-09-15 Boehringer Ingelheim Pharmaceuticals, Inc. Glucocorticoid mimetics, methods of making them, pharmaceutical compositions and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2136634A4 *

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10624882B2 (en) 2006-09-20 2020-04-21 Aerie Pharmaceuticals, Inc. Rho kinase inhibitors
US8030340B2 (en) 2006-11-23 2011-10-04 Astrazeneca Ab Indazolyl sulphonamide derivatives useful as glucocorticoid modulators
US7728030B2 (en) 2006-12-21 2010-06-01 Astrazeneca Ab Chemical compounds 572
US8143290B2 (en) 2006-12-21 2012-03-27 Astrazeneca Ab Chemical compounds 572
US9890123B2 (en) 2007-01-10 2018-02-13 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US10899714B2 (en) 2007-01-10 2021-01-26 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US9365518B2 (en) 2007-01-10 2016-06-14 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US8921392B2 (en) 2007-01-10 2014-12-30 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US10472327B2 (en) 2007-01-10 2019-11-12 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
US20100144713A1 (en) * 2008-01-17 2010-06-10 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
US8871757B2 (en) * 2008-01-17 2014-10-28 Aerie Pharmaceuticals, Inc. 6-and 7-amino isoquinoline compounds and methods for making and using the same
US8211930B2 (en) 2008-05-20 2012-07-03 Astrazeneca Ab Phenyl and benzodioxinyl substituted indazoles derivatives
US9738632B2 (en) 2008-05-20 2017-08-22 Astrazeneca Ab Phenyl and benzodioxinyl substituted indazoles derivatives
US8916600B2 (en) 2008-05-20 2014-12-23 Astrazeneca Ab Phenyl and benzodioxinyl substituted indazoles derivatives
US9512110B2 (en) 2008-05-20 2016-12-06 Astrazeneca Ab Phenyl and benzodioxinyl substituted indazoles derivatives
US10112920B2 (en) 2008-07-25 2018-10-30 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US10532993B2 (en) 2008-07-25 2020-01-14 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US9096569B2 (en) 2008-07-25 2015-08-04 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US10882840B2 (en) 2008-07-25 2021-01-05 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US11021456B2 (en) 2008-07-25 2021-06-01 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US9512101B2 (en) 2008-07-25 2016-12-06 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US9884840B2 (en) 2008-07-25 2018-02-06 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
US9951059B2 (en) 2009-05-01 2018-04-24 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US11028081B2 (en) 2009-05-01 2021-06-08 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US11618748B2 (en) 2009-05-01 2023-04-04 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US10654844B2 (en) 2009-05-01 2020-05-19 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US10316029B2 (en) 2009-05-01 2019-06-11 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
US10174017B2 (en) 2009-05-01 2019-01-08 Aerie Pharmaceuticals, Inc. Dual mechanism inhibitors for the treatment of disease
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
US11197853B2 (en) 2013-03-15 2021-12-14 Aerie Pharmaceuticals, Inc. Combination therapy
US11185538B2 (en) 2013-03-15 2021-11-30 Aerie Pharmaceuticals, Inc. Compositions for treating glaucoma or reducing intraocular pressure
US9415043B2 (en) 2013-03-15 2016-08-16 Aerie Pharmaceuticals, Inc. Combination therapy
US9993470B2 (en) 2013-03-15 2018-06-12 Aerie Pharmaceuticals, Inc. Combination therapy
US10568878B2 (en) 2013-03-15 2020-02-25 Aerie Pharmaceuticals, Inc. Combination therapy
US10588901B2 (en) 2013-03-15 2020-03-17 Aerie Pharmaceuticals, Inc. Combination therapy
US9931336B2 (en) 2013-03-15 2018-04-03 Aerie Pharmaceuticals, Inc. Combination therapy
US11020385B2 (en) 2013-03-15 2021-06-01 Aerie Pharmaceuticals, Inc. Combination therapy
US9849122B2 (en) 2013-03-15 2017-12-26 Aerie Pharmaceuticals, Inc. Combination therapy
US10550087B2 (en) 2015-11-17 2020-02-04 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US9643927B1 (en) 2015-11-17 2017-05-09 Aerie Pharmaceuticals, Inc. Process for the preparation of kinase inhibitors and intermediates thereof
US11389441B2 (en) 2016-08-31 2022-07-19 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
US11590123B2 (en) 2016-08-31 2023-02-28 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
US11707460B2 (en) 2016-08-31 2023-07-25 Aerie Pharmaceuticals, Inc. Ophthalmic compositions
US10858339B2 (en) 2017-03-31 2020-12-08 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US11312700B2 (en) 2017-03-31 2022-04-26 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US11427563B2 (en) 2018-09-14 2022-08-30 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds
US11891376B2 (en) 2018-09-14 2024-02-06 Aerie Pharmaceuticals, Inc. Aryl cyclopropyl-amino-isoquinolinyl amide compounds

Also Published As

Publication number Publication date
EP2136634A1 (en) 2009-12-30
US20100063282A1 (en) 2010-03-11
JP2010523689A (en) 2010-07-15
EP2136634A4 (en) 2011-07-06
CA2683647A1 (en) 2008-10-16

Similar Documents

Publication Publication Date Title
US7741361B2 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US20100063282A1 (en) Glucocorticoid Mimetics, Methods of Making Them, Pharmaceutical Compositions, and Uses Thereof
EP1539141B1 (en) 3-(sulfonamidoethyl)-indole derivatives for use as glucocorticoid mimetics in the treatment of inflammatory, allergic and proliferative diseases
US6858627B2 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US20100317863A1 (en) Glucocorticoid Mimetics, Methods of Making Them, Pharmaceutical Compositions and Uses Thereof
US7186864B2 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US20040010148A1 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US20040224992A1 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
US20050234091A1 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions and uses thereof
CA2558023A1 (en) 2-propanol and 2-propylamine derivatives and their use as glucocorticoid ligands
US20100048950A1 (en) Glucocorticoid Mimetics, Methods of Making Them, Pharmaceutical Compositions and Uses Thereof
US20090176807A1 (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof

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: 08745203

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12520997

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2008745203

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2010503136

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2683647

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE