US20100324093A1 - Ccr9 inhibitors and methods of use thereof - Google Patents

Ccr9 inhibitors and methods of use thereof Download PDF

Info

Publication number
US20100324093A1
US20100324093A1 US12/869,070 US86907010A US2010324093A1 US 20100324093 A1 US20100324093 A1 US 20100324093A1 US 86907010 A US86907010 A US 86907010A US 2010324093 A1 US2010324093 A1 US 2010324093A1
Authority
US
United States
Prior art keywords
benzenesulfonamide
chloro
phenyl
benzoyl
isopropoxy
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/869,070
Inventor
Paul Fleming
Geraldine C.B. Harriman
Zhan Shi
Shaowu Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Millennium Pharmaceuticals Inc
Original Assignee
Millennium Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Millennium Pharmaceuticals Inc filed Critical Millennium Pharmaceuticals Inc
Priority to US12/869,070 priority Critical patent/US20100324093A1/en
Publication of US20100324093A1 publication Critical patent/US20100324093A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/22Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms
    • C07C311/29Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic 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
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • 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
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/14Drugs for genital or sexual disorders; Contraceptives for lactation disorders, e.g. galactorrhoea
    • 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
    • A61P17/04Antipruritics
    • 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
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/15Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C311/21Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/30Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/37Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • C07C311/38Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring having sulfur atoms of sulfonamide groups and amino groups bound to carbon atoms of six-membered rings of the same carbon skeleton
    • C07C311/44Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring having sulfur atoms of sulfonamide groups and amino groups bound to carbon atoms of six-membered rings of the same carbon skeleton having the nitrogen atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/30Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/45Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound nitrogen atoms, not being part of nitro or nitroso groups at least one of the singly-bound nitrogen atoms being part of any of the groups, X being a hetero atom, Y being any atom, e.g. N-acylaminosulfonamides
    • C07C311/46Y being a hydrogen or a carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/26Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C317/32Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton with sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C317/34Sulfones; Sulfoxides having sulfone or sulfoxide groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton with sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having sulfone or sulfoxide groups and amino groups bound to carbon atoms of six-membered aromatic rings being part of the same non-condensed ring or of a condensed ring system containing that ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/46Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having at least one of the nitrogen atoms, not being part of nitro or nitroso groups, further bound to other hetero atoms
    • C07C323/49Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having at least one of the nitrogen atoms, not being part of nitro or nitroso groups, further bound to other hetero atoms to sulfur 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
    • C07D213/24Heterocyclic 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 substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/44Radicals substituted by doubly-bound oxygen, sulfur, or nitrogen atoms, or by two such atoms singly-bound to the same carbon atom
    • C07D213/46Oxygen 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
    • C07D213/24Heterocyclic 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 substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/44Radicals substituted by doubly-bound oxygen, sulfur, or nitrogen atoms, or by two such atoms singly-bound to the same carbon atom
    • C07D213/46Oxygen atoms
    • C07D213/50Ketonic radicals
    • 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
    • C07D213/63One oxygen atom
    • C07D213/65One oxygen atom attached in position 3 or 5
    • 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • C07D213/71Sulfur atoms to which a second hetero atom is attached
    • 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • 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
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • 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
    • C07D213/79Acids; Esters
    • C07D213/80Acids; Esters in position 3
    • 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
    • C07D213/89Heterocyclic 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 with hetero atoms directly attached to the ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/08Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon 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
    • C07D239/26Heterocyclic 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 only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon 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
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur 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
    • C07D333/30Hetero atoms other than halogen
    • C07D333/34Sulfur atoms
    • 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
    • C07D333/62Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • 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
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • Chemokines are a large and growing family of nearly forty 6-14 kD (non-glycosylated) heparin binding proteins that mediate a wide range of biological functions (Taub, D. D. and Openheim, J. J., Ther. Immunol., 1:229-246 (1994)).
  • the chemokines can be divided into families based on the position of four cysteine residues that form two disulfide bonds (Kelner, G. S., et al., Science, 266:12395-1399 (1994); Bazan, J. F., et al., Nature, 385:640-644 (1997); Pin, Y., et al., Nature, 385:611-617 (1997)).
  • Chemokine receptors can also be divided into families based on the type of chemokine they bind, although, no clear structural differences have been identified that distinguish the receptor sub-families (Mackay, C. R., J. Exp. Med., 184:799-802 (1996)).
  • Chemokines play a vital role in leukocyte adhesion and extravasation.
  • chemokines can induce the chemotaxis or transendothelial migration of leukocytes (Taub, D. D. and Openheim, J. J., Ther. Immunol., 1:229-246 (1994)), while in vivo injection (Taub, D. D., et al., J. Clin. Invest., 97:1931-1941 (1996)) or over-expression of chemokines (Fuentes, M. E., et al., J.
  • Immunol., 155:5769-5776 (1995) can result in leukocyte accumulation at the site of chemokine injection or expression.
  • Antagonists of chemokines can prevent leukocyte trafficking (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993)) and may have beneficial effects on acute and chronic inflammation (Sekido, N., et al., Nature, 365:654-657 (1993); Karpus, W. J., et al., J. Immunol., 155:5003-5010 (1995)).
  • Chemokines have also been reported to modulate angiogenesis (Gupta, S. K., et al., Proc.
  • chemokine receptors act as co-receptors, along with CD4, for entry of M tropic and T tropic HIV-1 (Choe, H., et al., Cell, 85:1135-1148 (1996); Feng, Y., et al., Science, 272:872-877 (1996)).
  • CD4 lymphocytes can be defined based on their expression of various adhesion molecules that are known to effect trafficking to different physiologic sites (Mackay, C. R., Curr. Opin. Immunol., 5:423-427 (1993)).
  • CLA +ve memory CD4 lymphocytes traffic to the skin (Berg, E. L., et al., Nature, 174(6):1461-1466 (1991)), while CLA ⁇ ve ⁇ 4 ⁇ 7 +ve memory CD4 lymphocytes traffic to mucosal sites (Hamman, A., et al., J. Immunol., 152:3282-3292 (1994)).
  • Leukocyte adhesion to endothelium is thought to involve several overlapping steps including rolling, activation and arrest. Rolling leukocytes are exposed to factors expressed at the adhesion site resulting in activation of the leukocyte and up-regulation of integrin-mediated adhesion. As a consequence of such integrin-mediated interactions, leukocytes arrest on the endothelium (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993); Bargatze, R. F., et al., Immunity, 3:99-108 (1995)).
  • Leukocyte activation and up-regulation of integrin molecules occurs via a pertussis toxin sensitive mechanism that is thought to involve chemokine receptors (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993); Campbell, J. J., et al., Science, 279:381-383 (1998)).
  • Memory CD4 + lymphocytes can be grouped based upon the expression of certain chemokine receptors.
  • CXCR3, CCR2 and CCR5 are all expressed on subsets of memory CD4 lymphocytes, and certain chemokines act selectively on naive T cells (Adema, G. J., et al., Nature, 387:713-717 (1997)).
  • chemokines which are ligands for such receptors have been shown to be expressed in inflammatory sites (Gonzalo, J. A., et al., J. Clin. Invest., 98:2332-2345 (1996)) and in some cases in lymph nodes draining a challenged site (Tedla, N., et al., J. Immunol., 161:5663-5672 (1998)).
  • In vitro derived T H 1/T H 2 lymphocyte lines have also been shown to differentially express chemokine receptors. Specifically, T H 1 lymphocytes have been shown to selectively express CXCR3 and CCR5, while T H 2 lymphocytes selectively express CCR4, CCR8 and CCR3 (Bonecchi, R.
  • chemokines for these respective chemokine receptors are induced by cytokines associated with a T H 1/T H 2 environment (Andrew, D. P., et al., J. Immunol. 161:5027-5038 (1998); Luster, A. D., et al., Nature, 315:672-676 (1985)).
  • the present invention relates to compounds represented by Structural Formula I, and pharmaceutically acceptable salts, solvates, and hydrates of such compounds:
  • Y is C(O), O, S, S(O) or S(O) 2 ;
  • X 1 , X 2 , and X 3 are each, independently, N or CR, provided that at least one of X 1 , X 2 , or X 3 is CR;
  • R for each occurrence, and R 1 are each, independently, H or a substituent.
  • the substituents at positions R and R 1 are each, independently, an aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, sulonamido, sulfone, sulfoxide, hydroxy, NR 11 CO 2 R 12 , C(O)N(R 11 ) 2 , C(O)R 12 , CO 2 R 12 , OC(O)N(R 11 ) 2 , OC(O)R 12 , N(R 11 ) 2 , or NR 11 C(O)R 12 .
  • R 11 and R 12 are defined further herein;
  • R 6 is H, an aliphatic carbonyl group, or an aliphatic ester; and
  • Ar 1 and Ar 2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group.
  • Ar 1 is a substituted or unsubstituted phenyl, a substituted or unsubstituted pyridyl, a substituted or unsubstituted pyrimidinyl, or a substituted or unsubstituted pyrazinyl.
  • Ar 2 is a substituted or unsubstituted phenyl, or a substituted or unsubstituted pyridyl. More preferably, Ar 1 is a substituted phenyl and Ar 2 is substituted pyridyl. Ring A is substituted or unsubstituted.
  • X 4 is N, N + —O ⁇ , or CR; and ring A, ring B, and ring C are each, independently, substituted or unsubstituted.
  • R is H or a substituent. Examples of substituents for R are defined as in Structural Formula I.
  • X 4 is N, N + —O ⁇ or CR; ring A, ring B, and ring C are each, independently, substituted or unsubstituted, and R is H or a substituent.
  • substituents for R are defined as in Structural Formula I; and R 8 is H or an electron withdrawing group.
  • R 8 is a halo, nitro, alkylcarbonyl or trihaloalkyl. More preferably, R 8 is Cl, Br or NO 2 .
  • compounds of the invention are represented by Structural Formula IV.
  • Ar 1 , Ar 2 , R 1 and R 6 are defined as in Structural Formula I; and R 19 and R 20 are each, independently, H or a substituent.
  • substituents in the R 19 and R 20 positions include an aliphatic group, a haloalkyl group, an ester, an amide, alkylcarbonyl, a halogen, COOH, NO 2 , alkoxy, haloalkoxy, CN, amino, and aminoalkyl.
  • compounds of the invention are represented by Structural Formula V.
  • X 4 is CR, N or N + —O ⁇ ;
  • X 1 , X 2 , X 3 , R, and Ar 2 are defined as in Structural Formula I;
  • R 1 , R 2 , R 3 , R 4 , R 5 are, independently, H, an aliphatic group, a haloalkyl group, a halo, COOH, NO 2 , or an alkoxy, a haloalkoxy.
  • X 1 is defined as in Structural Formula I;
  • X 4 is defined as in Structural Formula V;
  • R 8 is H or an electron withdrawing group;
  • m and n are each, independently, 0 or an integer from 1 to 3;
  • each R 9 is, independently, aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR 11 CO 2 R 12 , C(O)N(R 11 ) 2 , C(O)R 12 , CO 2 R 12 , OC(O)N(R 11 ) 2 , OC(O)R 12 , N(R 11 ) 2 , or NR 11 C(O)R 12 ; or two adjacent R 9 groups taken together with the atoms to which they are attached form a fused, saturated,
  • compounds of the invention are represented by Structural Formula VII.
  • ring B is substituted or unsubstituted;
  • X 1 is defined as in Structural Formula I;
  • R 8 is defined as in Structural Formula VI;
  • p is 0 or an integer from 1-3; and each R 13 is, independently, a halo or a substituted or unsubstituted heteroaryl.
  • compounds of the invention are represented by Structural Formula VIII.
  • ring A, Ar 1 , Ar 2 , X 1 , R 1 , R 6 are defined as in Structural Formula I; R 19 and R 20 are defined as in Structural Formula IV; and Y 1 is S, O, S(O), or S(O) 2 .
  • the present invention provides a method of inhibiting a CCR9 receptor by contacting the receptor with an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof.
  • the invention provides a method of inhibiting CCR9-mediated homing of leukocytes in a subject by administering to the subject an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof.
  • the method inhibits homing of leukocytes to mucosal tissue.
  • the invention provides a method of treating a subject having a disease that is mediated by a CCR9 receptor, such as an inflammatory disease, for example Celiac's disease or an inflammatory bowel disease, by administering to the subject an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof.
  • a CCR9 receptor such as an inflammatory disease, for example Celiac's disease or an inflammatory bowel disease
  • the present invention provides a pharmaceutical composition having a pharmaceutically acceptable carrier and at least one compound of the present invention.
  • Chemokines and their associated receptors are proinflammatory mediators that promote recruitment and activation of multiple lineages of leukocytes and lymphocytes. Continuous release of chemokines at sites of inflammation mediates the ongoing migration of effector cells in chronic inflammation.
  • CCR9 and its associated chemokine TECK have been implicated in chronic inflammatory diseases, such as inflammatory bowel diseases.
  • Small molecule inhibitors of the interaction between CCR9 and its ligands e.g., TECK
  • these compounds of the invention are useful for inhibiting harmful inflammatory processes triggered by receptor-ligand interactions and thus are useful for treating diseases mediated by CCR9, such as chronic inflammatory diseases.
  • aliphatic as used herein means straight-chain, branched or cyclic C 1 -C 12 hydrocarbons which are completely saturated or which contain one or more units of unsaturation but which are not aromatic.
  • suitable aliphatic groups include substituted or unsubstituted linear, branched or cyclic alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • alkyl alkoxy
  • alkylthio used alone or as part of a larger moiety includes both straight and branched chains containing one to twelve carbon atoms.
  • alkenyl and alkynyl used alone or as part of a larger moiety shall include both straight and branched chains containing two to twelve carbon atoms.
  • cycloalkyl used alone or as part of a larger moiety shall include cyclic C 3 -C 12 hydrocarbons which are completely saturated or which contain one or more units of unsaturation, but which are not aromatic.
  • aryl groups are carbocyclic aromatic ring systems (e.g. phenyl), fused polycyclic aromatic ring systems (e.g. naphthyl and anthracenyl) and aromatic ring systems fused to carbocyclic non-aromatic ring systems (e.g., 1,2,3,4-tetrahydronaphthyl) having six to about fourteen carbon atoms.
  • carbocyclic aromatic ring systems e.g. phenyl
  • fused polycyclic aromatic ring systems e.g. naphthyl and anthracenyl
  • aromatic ring systems fused to carbocyclic non-aromatic ring systems e.g., 1,2,3,4-tetrahydronaphthyl having six to about fourteen carbon atoms.
  • haloalkyl means alkyl, alkenyl or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • halogen means F, Cl, Br or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to aromatic ring groups having five to fourteen members, such as phenyl, benzyl, phenethyl, 1-napthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl.
  • aryl also refers to rings that are optionally substituted.
  • aryl may be used interchangeably with the term “aryl ring”.
  • Aryl also includes fused polycyclic aromatic ring systems in which an aromatic ring is fused to one or more rings.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as in an indanyl, phenantriidinyl, or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic ring.
  • heteroatom means nitrogen, oxygen, or sulfur and includes any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen.
  • nitrogen includes a substitutable nitrogen of a heterocyclic ring.
  • the nitrogen in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR 18 (as in N-substituted pyrrolidinyl).
  • heterocycle includes non-aromatic ring systems having five to fourteen members, preferably five to ten, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, or S.
  • heterocyclic rings examples include 3-1H-benzimidazol-2-one, 3-tetrahydrofuranyl, 2-tetrahydropyranyl, 3-tetrahydropyranyl, 4-tetrahydropyranyl, [1,3]-dioxalanyl, [1,3]-dithiolanyl, [1,3]-dioxanyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-thiomorpholinyl, 3-thiomorpholinyl, 4-thiomorpholinyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrorolidinyl, 1-piperazinyl, 2-piperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substituted diazol
  • heterocycle is a group in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic or non-aromatic rings, such as in an indolinyl, chromanyl, phenantrhidinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring.
  • heterocycle whether saturated or partially unsaturated, also refers to rings that are optionally substituted.
  • heteroaryl used alone or as part of a larger moiety as in “heteroaralkyl” or “heteroarylalkoxy”, refers to heteroaromatic ring groups having five to fourteen members.
  • heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxadiazolyl, 5-oxadiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 3-pyridazinyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-triazoly
  • heteroaryl is a group in which a heteroatomic ring is fused to one or more aromatic or nonaromatic rings where the radical or point of attachment is on the heteroaromatic ring. Examples include tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[3,4-d]pyrimidinyl.
  • heteroaryl also refers to rings that are optionally substituted.
  • Preferred heteroaryl groups are thienyl, benzo(b)thienyl, pyrrolyl, indolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, furanyl, and benzo(b)furanyl. More preferred heteroaryl groups are pyridyl and thienyl.
  • arylalkyl group is an aryl substituent that is linked to a compound by an alkyl group having from one to twelve carbon atoms.
  • alkoxy group is a C 1 -C 12 alkyl group that is connected to a compound via an oxygen atom.
  • alkoxy groups include but are not limited to methoxy, ethoxy, propoxy, isopropoxy, and t-butoxy.
  • a cycloalkoxy group is a cyclic C 3 -C 12 hydrocarbon which is attached to a compound via an oxygen.
  • Cycloalkoxy groups include but are not limited to cyclopropoxy and cyclobutoxy.
  • a haloalkoxy is a haloalkyl group that is attached to a compound via an oxygen.
  • a preferred haloalkoxy is trifluoromethoxy.
  • aryloxy as used herein, is an aryl group that is attached to a compound via an oxygen.
  • a preferred aryloxy is phenoxy.
  • a arylalkoxy group is a arylalkyl group that is attached to a compound via an oxygen on the C 1 -C 12 alkyl portion of the arylalkyl.
  • a preferred arylalkoxy is phenylmethoxy.
  • alklythio group is a C 1 -C 12 alkyl group that is connected to a compound via a sulfur atom.
  • a aliphatic carbonyl group is an aliphatic group that is connected to a compound via a carbonyl group.
  • a preferred aliphatic carbonyl is acetyl.
  • a aliphatic ester group as used herein, an aliphatic group that is connected to a compound via an ester linkage (i.e., —C(O)O-aliphatic group).
  • An electron withdrawing group is a group which causes a dipole moment in the direction of the group.
  • Suitable electron withdrawing groups include but are not limited to halo (preferably chloro), haloalkyl (preferably trifluoromethyl), nitro, cyano, sulfonamido, sulfone, and sulfoxide.
  • An aryl including aralkyl, aralkoxy, aryloxyalkyl and the like
  • heteroaryl including heteroaralkyl and heteroarylalkoxy and the like
  • substituents include aliphatic groups, aryl groups, haloalkoxy groups, heteroaryl groups, halo, hydroxy, OR 14 , COR 14 , COOR 14 , NHCOR 14 , OCOR 14 , benzyl, haloalkyl (e.g., trifluoromethyl and trichloromethyl), cyano, nitro, S(O), S(O) 2 , SO 3 ⁇ , SH, SR 14 , NH 2 , NHR 14 , NR 14 R 15 , NR 6 S(O) 2 —R 7 , and COOH, wherein R 14 and R 15 are each, independently, an aliphatic group, a cycloalkyl, an aryl group, or
  • substituents for an aryl or heteroaryl group include —R 16 , —OR 16 , —SR 16 , 1,2-methylene-dioxy, 1,2-ethylenedioxy, protected OH (such as acyloxy), phenyl (Ph), substituted Ph, —O(Ph), substituted —O(Ph), —CH 2 (Ph), substituted —CH 2 CH 2 (Ph), substituted —CH 2 CH 2 (Ph), —N(R 16 ) 2 , —NR 16 CO 2 R 16 , —NR 16 NR 16 C(O)R 16 , —NR 16 R 16 C(O)N(R 16 ) 2 , —NR 16 NR 16 CO 2 R 16 , —C(O)C(O)R 16 , —C(O)CH 2 C(O)R 16 , —CO 2 R 16 , —C(O)R 16 , —C(O)N(R 16 ) 2 , —OC(
  • Preferred substituents on the aryl or heteroaryl group include halo, haloalkyl, sulfone, sulfoxide, nitro, cyano, alkyl group, alkoxy group, and alkylamino group.
  • substituents on the aliphatic group or the phenyl ring include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • Substituents for an aryl or a heteroaryl group include aryl groups (including a carbocyclic aryl group or a heteroaryl group), aliphatic groups, cycloalkyl groups, haloalkoxy groups, heteroaryl groups, hydroxy, OR 14 , aldehyde, COR 14 , COOR 14 , NHCOR 14 , OCOR 14 , benzyl, haloalkyl (e.g., trifluoromethyl and trichloromethyl), halo, cyano, nitro, SO 3 ⁇ , SH, SR 14 , NH 2 , NHR 14 , NR 14 R 15 , NR 6 S(O) 2 —R 7 , or COOH, wherein R 14 and R 15 are each, independently, an aliphatic group, a cycloalkyl, an aryl group, or an arylalky group.
  • An aliphatic group or a heterocycle may contain one or more substituents.
  • suitable substituents on the saturated carbon of an aliphatic group of a heterocycle include those listed above for an aryl or heteroaryl group and the following: ⁇ O, ⁇ S, ⁇ NNHR 17 , ⁇ NN(R 17 ) 2 , ⁇ NNHC(O)R 17 , ⁇ NNHCO 2 (alkyl), ⁇ NNHSO 2 (alkyl), or ⁇ NR 17 , where each R 17 is independently selected from hydrogen, an unsubstituted aliphatic group or a substituted aliphatic group.
  • substituents on the aliphatic group include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, thioalkyl, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • Suitable substitutents on the nitrogen of a non-aromatic heterocycle or on an unsaturated nitrogen of a heteroaryl include —R 18 , —N(R 18 ) 2 , —C(O)R 18 , —CO 2 R 18 , —C(O)C(O)R 18 , —C(O)CH 2 C(O)R 18 , —SO 2 R 18 , —SO 2 N(R 18 ) 2 , —C( ⁇ S)N(R 18 ) 2 , —C( ⁇ NH)—N(R 18 ) 2 , and —NR 18 SO 2 R 18 ; wherein R 18 is hydrogen, an aliphatic group, a substituted aliphatic group, phenyl (Ph), substituted Ph, —O(Ph), substituted —O(Ph), —CH 2 (Ph), or an unsubstituted heteroaryl or heterocyclic ring.
  • substituents on the aliphatic group or the phenyl ring include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • linker group means an organic moiety that connects two parts of a compound.
  • Linkers are typically comprised of an atom, such as oxygen or sulfur, a unit, such as —NH—, —CH 2 —, —C(O)—, or —C(O)NH—, or a chain of atoms, such as an alkylene chain.
  • the molecular mass of a linker is typically in the range of about 14 to 200, preferably in the range of 14 to 96 with a length of up to about six atoms.
  • linkers include a saturated or unsaturated C 1 -C 6 alkylene chain which is substituted or unsubstituted, and wherein one or two saturated carbons of the chain are optionally replaced by —C(O)—, —C(O)C(O)—, —CONH—, —CONHNH—, —CO 2 —, —OC(O)—, —NHCO 2 —, —O—, —NHCONH—, —OC(O)NH—, —NHNH—, —NHCO—, —S—, —SO—, —SO 2 —, —NH—, —SO 2 NH—, or —NHSO 2 —.
  • the compounds of the present invention have IC50s of less than 1 ⁇ m; less than 750 nm; preferably less than 500 nm; more preferably less than 250 nm; even more preferably less than 100 nm, most preferably less than 50 nm; and the most preferable less than 10 nm, e.g., less than 5 nm.
  • mammalian CCR9 refers to naturally occurring or endogenous mammalian CCR9 proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian CCR9 protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature receptor protein, polymorphic or allelic variants, and other isoforms of a mammalian CCR9 (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated, unglycosylated).
  • Naturally occurring or endogenous mammalian CCR9 proteins include wild type proteins such as mature CCR9, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces mammalian CCR9, for example.
  • Polymorphic, allelic, splice and other naturally occurring variants of mammalian CCR9 can be expressed in particular organs, tissues or cells and have altered properties (e.g., altered affinity for ligand (e.g. TECK)) and specialized biological function (e.g., T cell development, T cell recruitment).
  • Naturally occurring or endogenous mammalian CCR9 proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding mammalian CCR9 are referred to by the name of the corresponding mammal.
  • the protein is designated as a human CCR9 protein (e.g., a recombinant human CCR9 produced in a suitable host cell).
  • mammalian TECK refers to naturally occurring or endogenous mammalian TECK proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian TECK protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature receptor protein, polymorphic or allelic variants, and other isoforms of a mammalian TECK (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated, unglycosylated).
  • Naturally occurring or endogenous mammalian TECK proteins include wild type proteins such as mature TECK, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces mammalian TECK, for example.
  • pharmaceutically acceptable means that the carrier, diluent, excipients and salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • Pharmaceutical formulations of the present invention are prepared by procedures known in the art using well known and readily available ingredients.
  • Preventing refers to reducing the likelihood that the recipient will incur or develop any of the pathological conditions described herein, for example inhibiting the onset of these symptoms.
  • Treating refers to eliminating or reducing the severity of a pathological condition described herein and/or eliminating, alleviating and/or reducing the symptoms of a pathological condition described herein.
  • the method is a method of inhibiting a CCR9 function in a subject.
  • the method is a method of treating a subject having an inflammatory disease associated with mucosal tissue, such as Crohn's disease, colitis, or Celiac disease.
  • the method is a method of inhibiting CCR9-mediated homing of leukocytes in a subject. See Papadakis, et al., Gastroenterology (2001), 121:246-254 for a discussion of the relationship of CCR9 receptors to the above diseases or conditions.
  • the compounds of the invention are represented by any one of Structural Formulas IX-XXIV:
  • Structural Formulas IX-XXV, Ar 2 and Y are defined as in Structural Formula I; and rings A and B are substituted or unsubstituted.
  • R 8 is defined as in Structural Formula V, and R 13 and p are defined as in Structural Formula VII.
  • Y in Structural Formula I, or IX-XXIV is C ⁇ O.
  • ring C in Structural Formula II or III or Ar 2 in Structural Formula I, IV, V or in any one of Structural Formulas IX-XXV is unsubstituted or substituted with one or more substituents selected from aliphatic group (including substituted aliphatic groups such as haloalkyl) aryl, arylalkyl, alkoxy (including cycloalkoxy and substituted alkoxy groups such as haloalkoxy), aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, S(O)-(aliphatic), S(O) 2 -(aliphatic), NR 11 S(O) 2 -(aliphatic), C(O)N(R 11 ) 2 , C(O)R 12 , N(R 11 ) 2 , NR 11 C(O) 2 R 12 and NR 11 C(O)R 12 , wherein R 11 for each substituents selected from ali
  • ring A in Structural Formula I or IV, or one or both rings A and B in Structural Formula II or III or in any one of Structural Formulas IX-XXV are, independently, substituted with a substituent selected from halo, aliphatic group, alkoxy, and haloalkyl.
  • ring A in Structural Formula I, II, III, IV, V, VI, VII, VIII, or in any one of Structural Formulas IX-XXV is substituted with an electron withdrawing substituent para to the sulfonamide group.
  • X 3 in Structural Formula V is CR 21 , wherein R 21 is an electron withdrawing group. More preferably, R 21 is halo, nitro, aliphatic carbonyl, or trihalomethyl. Most preferably, R 21 is Cl, Br, or nitro.
  • Ar 2 in Structural Formula I, IV, V, VIII or in any one of Structural Formulas IX-XXV is a substituted or unsubstituted phenyl, a substituted or unsubstituted naphthyl, a substituted or unsubstituted thienyl, or a substituted or unsubstituted thianaphthenyl. More preferably, Ar 2 is a substituted or unsubstituted phenyl or a substituted or unsubstituted thienyl.
  • R 9 for each occurrence in Structural Formula VI is, independently, an aliphatic group, an alkoxy, or a haloalkoxy.
  • R 8 in Structural Formula I, VI, VII, VIII, XXV and XXVI is a halo, nitro, alkylcarbonyl or trihaloalkyl. More preferably, R 8 is Cl, Br or NO 2 .
  • Y and Ar 2 in Structural Formula I and VIII or in any one of Structural Formulas IX-XXV are defined as in the first and the sixth preferred embodiments, respectively.
  • Ar 2 Structural Formula V is defined as in the sixth preferred embodiments, and X 3 is defined as in the fifth preferred embodiment.
  • X 4 in Structural Formulas V or VI is an nitrogen oxide (N + —O ⁇ ).
  • n is one in Structural Formula VI and R 9 is para to the sulfonamide substituent. More preferably, R 9 is as defined in seventh preferred embodiment.
  • m is one in Structural Formula VI and R 10 is meta to the carbonyl substituent.
  • R 6 in Structural Formula I, IV, or VIII is H.
  • Examples 7-27, 31-36, and 116-119 of Table 1 are shown in the Examples section.
  • Examples 45-143 (Examples 116-119 are also included below) of Table 1 have the following structural formulas:
  • chemokine and compound of the invention can be monitored (see e.g., Van i, J. et al., J. Exp. Med., 176: 59-65 (1992); Zachariae, C. O. C. et al., J. Exp. Med. 171: 2177-2182 (1990); Jose, P. J. et al., J. Exp. Med.
  • skin biopsies are assessed histologically for infiltration of leukocytes (e.g., CCR9 + T cells).
  • labeled cells e.g., stably transfected cells expressing a mammalian CCR9, labeled with 111 In for example
  • compound of the invention to be assessed which binds a mammalian CCR9 can be administered, either before, simultaneously with or after a CCR9 ligand or agonist (e.g., TECK) is administered to the test animal.
  • TECK CCR9 ligand or agonist
  • CCR9 is selectively expressed on memory lymphocytes which home to mucosal sites (e.g., CLA ⁇ ve ⁇ 4 ⁇ 7 hi CD4 + lymphocytes).
  • mucosal sites e.g., CLA ⁇ ve ⁇ 4 ⁇ 7 hi CD4 + lymphocytes.
  • animal models of inflammatory diseases of the mucosa e.g., respiratory tract, urogenital tract, alimentary canal and associated organs and tissues (e.g., pancreas, liver, gall bladder)
  • the therapeutic efficacy of a compound of the invention can be studied in the cotton-top tamarin model of inflammatory bowel disease (Podolsky, D. K., et al., J. Clin. Invest. 92:372-380 (1993)).
  • the CD45RB Hi /SCID model provides a mouse model with similarity to both Crohn's disease and ulcerative colitis (Powrie, F. et al., Immunity, 1: 553-562 (1994)).
  • Therapeutic efficacy in this model can be assessed, for example, by using parameters such as inhibition of recruitment of 111 In-labeled cells to the colon and reduction in the number of CD4 + T lymphocytes in the lamina intestinal of the large intestine after administration (e.g., intravenous (i.v.), intraperitoneally (i.p.) and per oral (p.o.)) of a compound.
  • Inhibition of at least one function characteristic of a mammalian CCR9 protein according to the present invention provides an effective and selective way of inhibiting receptor-mediated functions. Once lymphocytes are recruited to a site, other leukocyte types, such as monocytes, may be recruited by secondary signals. Thus, compounds which can inhibit CCR9 function (e.g., compounds of the invention) can be used to inhibit leukocyte function (e.g., leukocyte infiltration including recruitment and/or accumulation).
  • the present invention provides a method of inhibiting an inflammatory response in a subject, comprising administering an effective amount of a compound of the invention which inhibits mammalian CCR9 function to a subject in need of such therapy.
  • an effective amount of a compound which inhibits one or more functions of a mammalian CCR9 protein e.g., a human CCR9
  • a mammalian CCR9 protein e.g., a human CCR9
  • Preferred compounds of the invention inhibit an inflammatory response in a subject by inhibiting (i.e., reduce or prevent) binding of ligand (e.g. TECK) to CCR9.
  • one or more inflammatory processes such as leukocyte emigration, chemotaxis, exocytosis (e.g., of enzymes) or inflammatory mediator release, is inhibited.
  • leukocytic infiltration of inflammatory sites e.g., in a inflamed mucus membrane (e.g., colon, small intestine)
  • an effective amount of a compound of the invention which inhibits one or more functions of a mammalian CCR9 protein e.g., a human CCR9
  • an effective amount of a compound which binds to human CCR9 and/or an effective amount of a compound which binds to human TECK is administered to a subject in need thereof.
  • the invention relates to a method of treating a subject having an inflammatory disease, comprising administering an effective amount of a compound of the invention that antagonizes CCR9 function.
  • the subject has an inflammatory bowel disease, such as Crohn's disease or colitis.
  • Treatment includes therapeutic or prophylactic treatment.
  • Treatment in accordance with the method, can prevent disease or reduce the severity of disease in whole or in part.
  • the invention also relates to a method of inhibiting CCR9-mediated homing of leukocytes in a subject, comprising administering an effective amount of a compound of the invention that antagonizes CCR9 function, for example, the homing of leukocytes to mucosal sites can be inhibited.
  • a compound of the invention that antagonizes CCR9 function
  • the homing of leukocytes to mucosal sites can be inhibited.
  • Immigration of circulating leukocytes into organs or tissue (e.g., intestine) and/or local recruitment of lymphocytes within an organ or tissue e.g., IEL, LPL
  • subject is defined herein to include animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent or murine species.
  • a “subject in need of treatment to inhibit CCR9 function” is a subject in whom a beneficial therapeutic or prophylactic effect can be achieved by inhibiting CCR9 function. Examples include subjects with one of the diseases or conditions described herein.
  • the disease or condition is one in which the actions of lymphocytes, particularly lymphocytes which home to mucosal tissues, are to be inhibited or promoted for therapeutic (including prophylactic) purposes.
  • the inflammatory disease or condition is a T cell-mediated disease or condition.
  • inflammatory diseases associated with mucosal tissues include mastitis (mammary gland), vaginitis, cholecystitis, cholangitis or pericholangitis (bile duct and surrounding tissue of the liver), chronic bronchitis, chronic sinusitis, asthma, and graft versus host disease (e.g., in the gastrointestinal tract).
  • interstitial fibrosis e.g., interstitial lung diseases (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with rheumatoid arthritis, or other autoimmune conditions), hypersensitivity pneumonitis, collagen diseases, sarcoidosis, and other idiopathic conditions can be amenable to treatment.
  • ILD interstitial lung diseases
  • Pancreatitis and insulin-dependent diabetes mellitus are other diseases which can be treated using the present method.
  • diseases which can be treated accordingly include inflammatory bowel disease (IBD), such as ulcerative colitis, Crohn's disease, ileitis, Celiac disease, nontropical Sprue, enteritis, enteropathy associated with seronegative arthropathies, microscopic or collagenous colitis, eosinophilic gastroenteritis, or pouchitis resulting after proctocolectomy, and ileoanal anastomosis.
  • IBD inflammatory bowel disease
  • Additional diseases or conditions including chronic diseases, of humans or other species which can be treated with compounds of the invention that inhibit of CCR9 function, include, but are not limited to:
  • the compound can be administered as a neutral compound or as a salt.
  • Salts of compounds containing an amine or other basic group can be obtained, for example, by contacting the compound with a suitable organic or inorganic acid, such as hydrogen chloride, hydrogen bromide, acetic acid, perchloric acid and the like.
  • Compounds with a quaternary ammonium group also contain a counteranion such as chloride, bromide, iodide, acetate, perchlorate and the like.
  • Other examples of such salts include hydrochlorides, hydrobromides, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, tartrates [e.g. (+)-tartrates, ( ⁇ )-tartrates or mixtures thereof including racemic mixtures], succinates, benzoates and salts with amino acids such as glutamic acid.
  • Salts of compounds containing a carboxylic acid or other acidic functional group can be prepared by reacting with a suitable base.
  • a suitable base which affords a pharmaceutically acceptable cation, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and magnesium), aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N,N′-dibenzylethylenediamine, 2-hydroxyethylamine, bis-(2-hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine, N-benzyl- ⁇ -phenethylamine, dehydroabietylamine, N,N′-bisdehydroabietylamine, glucamine, N-methylglucamine, coll
  • Salts of acidic functional groups contain a counteraction such as sodium, potassium and the like.
  • Certain compounds described herein and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
  • Certain compounds described herein may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers.
  • the present invention includes each conformational isomer of these compounds and mixtures thereof.
  • Certain compounds described herein may exist in zwitterionic form.
  • the present invention includes each zwitterionic form of these compounds and mixtures thereof.
  • Certain compounds described herein and their salts may exist in more than one crystal form.
  • Polymorphs of these compounds form part of this invention and may be prepared by crystallization of the compound under different conditions, for example, by using different solvents or different solvent mixtures for recrystallization; by crystallization at different temperatures; or by various modes of cooling, ranging from very fast to very slow cooling during crystallization. Polymorphs may also be obtained by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs may be determined by solid probe nmr spectroscopy, it spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques.
  • an “effective amount” or “pharmaceutically effective amount” is intended to include an amount which is sufficient to ameliorate a disease or condition and prevent its further progression or ameliorate the symptoms associated with the disease or condition. Such an amount can be administered prophylactically to a patient thought to be susceptible to development of a disease or condition. Such amount when administered prophylactically to a patient can also be effective to prevent or lessen the severity of the mediated condition. Such an amount is intended to include an amount which is sufficient to modulate a CCR9 receptor-mediated disease or condition. Conditions mediated by CCR9 receptors include all of the diseases or conditions described herein.
  • an “effective amount” typically ranges between about 0.01 mg/kg/day to about 100 mg/kg/day, preferably between about 0.5 mg/kg/day to about 50 mg/kg/day.
  • the compounds described herein, and the pharmaceutically acceptable salts, solvates and hydrates thereof, have valuable pharmacological properties and can be used in pharmaceutical preparations containing the compound or pharmaceutically acceptable salts thereof, in combination with a pharmaceutically acceptable carrier or diluent. They are useful as therapeutic substances in preventing or treating diseases mediated by CCR9 receptors, such as inflammatory diseases, in human or non-human animals.
  • Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions.
  • the active compound will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein. Techniques for formulation and administration of the compounds of the instant invention can be found in Remington: the Science and Practice of Pharmacy, 19 th edition, Mack Publishing Co., Easton, Pa. (1995).
  • the compound or salts thereof can be combined with a suitable solid or liquid carrier or diluent to form capsules, tablets, pills, powders, syrups, solutions, suspensions and the like.
  • the tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacias, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid, a lubricant such as magnesium stearate; and a sweetening agent such as sucrose lactose or saccharin.
  • a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • tablets may be coated with shellac, sugar or both.
  • a syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
  • the active compounds can also be administered intranasally as, for example, liquid drops or spray.
  • the compounds for use according to the present invention are conveniently delivered in the form of a dry powder inhaler, or an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or
  • the compounds of the present invention, or salts thereof can be combined with sterile aqueous or organic media to form injectable solutions or suspensions.
  • aqueous or organic media for example, solutions in sesame or peanut oil, aqueous propylene glycol and the like can be used, as well as aqueous solutions of water-soluble pharmaceutically-acceptable salts of the compounds.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that each syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against any contamination.
  • the carrier can be solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • the injectable solutions prepared in this manner can then be administered intravenously, intraperitoneally, subcutaneously, or intramuscularly, with intramuscular administration being preferred in humans.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation.
  • Such long acting formulations may be administered by implantation, for example, subcutaneously or intramuscularly or by intramuscular injection.
  • implantation for example, subcutaneously or intramuscularly or by intramuscular injection.
  • sparingly soluble derivatives for example, as sparingly soluble salts.
  • the effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated.
  • compositions are formulated and administered in the same general manner as detailed herein.
  • the compounds of the instant invention may be used effectively alone or in combination with one or more additional active agents depending on the desired target therapy.
  • Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound described herein and one or more additional active agents, as well as administration of the compound and each active agent in its own separate pharmaceutical dosage formulation.
  • a compound described herein or salt thereof can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations.
  • a compound of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
  • An effective amount of the compounds described herein can be used for the preparation of a medicament useful for treating a disease mediated by CCR9 receptors, such as an inflammatory disease, and for treating, preventing or reducing the risk of developing a disease mediated by CCR9 receptors, such as an inflammatory disease, in mammals, particularly in humans.
  • Preferably compounds of the invention or pharmaceutical formulations containing these compounds are in unit dosage form for administration to a mammal.
  • the unit dosage form can be any unit dosage form known in the art including, for example, a capsule, an IV bag, a tablet, or a vial.
  • the quantity of active ingredient (viz., a compound of Structural Formula I or salts thereof) in a unit dose of composition is an effective amount and may be varied according to the particular treatment involved. It may be appreciated that it may be necessary to make routine variations to the dosage depending on the age and condition of the patient.
  • the dosage will also depend on the route of administration which may be by a variety of routes including oral, aerosol, rectal, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal and intranasal.
  • compositions of the invention are prepared by combining (e.g., mixing) an effective amount of a compound of the invention together with a pharmaceutically acceptable carrier or diluent.
  • a pharmaceutically acceptable carrier or diluent e.g., a pharmaceutically acceptable carrier or diluent.
  • present pharmaceutical formulations are prepared by known procedures using well known and readily available ingredients.
  • the active ingredient will usually be admixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a capsule, sachet, paper or other container.
  • a carrier which may be in the form of a capsule, sachet, paper or other container.
  • the carrier serves as a diluent, it may be a solid, lyophilized solid or paste, semi-solid, or liquid material which acts as a vehicle, or can be in the form of tablets, pills, powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), or ointment, containing, for example, up to 10% by weight of the active compound.
  • the compounds of the present invention are preferably formulated prior to administration.
  • the carrier may be a solid, liquid, or mixture of a solid and a liquid.
  • compositions containing the compounds of the invention or the salts thereof may be provided in dosage unit form, each dosage unit containing from about 5% to about 95%, preferably about 20% to about 80%, of a compound of the invention, although it will, of course, readily be understood that the amount of the compound or compounds actually to be administered will be determined by a physician, in the light of all the relevant circumstances.
  • Powders and tablets preferably contain from about 1 to about 99 weight percent of the active ingredient which is the novel compound of this invention.
  • Suitable solid carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, low melting waxes, and cocoa butter.
  • Active Ingredient refers to a compound according to Structural Formula I or salts thereof.
  • Hard gelatin capsules are prepared using the following ingredients:
  • a tablet is prepared using the ingredients below:
  • the components are blended and compressed to form tablets each weighing 665 mg
  • An aerosol solution is prepared containing the following components:
  • the Active Ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 30° C. and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remainder of the propellant. The valve units are then fitted to the container.
  • Tablets each containing 60 mg of Active ingredient, are made as follows:
  • the Active Ingredient, starch and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly.
  • the aqueous solution containing polyvinylpyrrolidone is mixed with the resultant powder, and the mixture then is passed through a No. 14 mesh U.S. sieve.
  • the granules so produced are dried at 50° C. and passed through a No. 18 mesh U.S. sieve.
  • the sodium carboxymethyl starch, magnesium stearate and talc, previously passed through a No. 60 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
  • Capsules each containing 80 mg of Active Ingredient, are made as follows:
  • the Active Ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 45 mesh U.S. sieve, and filled into hard gelatin capsules in 200 mg quantities.
  • Suppositories each containing 225 mg of Active Ingredient, are made as follows:
  • the Active Ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool.
  • Suspensions each containing 50 mg of Active Ingredient per 5 mL dose, are made as follows:
  • Active Ingredient 50 mg Sodium carboxymethyl cellulose 50 mg Syrup 1.25 mL Benzoic acid solution 0.10 mL Flavor q.v. Color q.v. Purified water to total 5 mL
  • the Active Ingredient is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form a smooth paste.
  • the benzoic acid solution, flavor and color are diluted with a portion of the water and added, with stirring. Sufficient water is then added to produce the required volume.
  • An intravenous formulation may be prepared as follows:
  • the solution of the above materials generally is administered intravenously to a subject at a rate of 1 mL per minute.
  • the sulfonamides of the invention can be synthesized by treating a protected aromatic amine (XXVI) or a protected heteroaromatic amine (XXVI), such as an aminobenzene, an aminopyridine, an aminopyrimidine, an aminopyridazine, an aminopyrazine, an aminotriazine or an aminotetrazine, with an alkyl lithium.
  • a protected aromatic amine (XXVI) or a protected heteroaromatic amine (XXVI) such as an aminobenzene, an aminopyridine, an aminopyrimidine, an aminopyridazine, an aminopyrazine, an aminotriazine or an aminotetrazine, with an alkyl lithium.
  • the amino group of the aromatic amine or heteroaromatic amine is protected with protecting group which is stable under basic condition, such as a tert-butoxycarbonyl (BOC) group.
  • BOC tert-butoxycarbonyl
  • alkyl lithium e.g., n-butyl lithium, s-butyl lithium or t-butyl lithium
  • alkyl lithium e.g., n-butyl lithium, s-butyl lithium or t-butyl lithium
  • the temperature of the reaction mixture is allowed to increase to about ⁇ 25° C. to about 25° C.
  • about 1 equ. to about 1.5 equ. of a Weinreb amide (XXVII) such as a N-methoxy-N-methyl-benzamide or a N-methoxy-N-methyl-isonicotinamide, dissolved in an aprotic solvent is added to the reaction mixture.
  • the reaction is allowed to continue stirring until the protected aromatic amine (XXVI) or protected heteroaromatic amine (XXVI) is consumed (typically, for about 0.5 hr to about 3 hr at about 15° C. to about 35° C.).
  • the reaction is monitored by thin layer chromatography (tic) to determine when the aromatic amine (XXVI) or heteroaromatic amine (XXVI) has been consumed.
  • tic thin layer chromatography
  • compound XXIX is converted to a sulfonamide by using one of two alternative methods.
  • method 1 compound XXIX is dissolved in an aprotic solvent and the solution is cooled to about ⁇ 25° C. to about 10° C. and about 1 equ. to about 1.5 equ. of NaH is added to the mixture.
  • XXX sulfonyl chloride
  • An additional, amount of up to about 0.5 equ. NaH may be added to the reaction mixture.
  • reaction is stirred at about 15° C. to about 30° C. until the reaction is complete as indicated by tlc (typically, about 1 hr to about 3 hr).
  • tlc typically, about 1 hr to about 3 hr.
  • the reaction is quench by addition of water to yield sulfonamide (XXXI).
  • compound XXIX is dissolved in an aprotic solvent and the mixture is cooled to about ⁇ 10° C. to about 10° C.
  • An excess amount of sodium bis(trimethylsilyl)amide typically, about 1.5 equ. to about 3 equ.
  • a solution of about 1 equ. to about 2 equ. of sulfonyl chloride (XXX) in an aprotic solvent is added to the reaction mixture and the reaction is allowed to warm up to about 15° C. to about 30° C. and is stirred for about 6 hr to about 24 hr.
  • the reaction is quenched with an acidic water solution to yield sulfamide (XXXI).
  • a substituted Weinreb amide (10 mmol) and a substituted bromobenzene (10 mmol) were dissolved in THF under nitrogen.
  • the reaction mixture was cooled to ⁇ 100° C. using a liquid nitrogen/diethyl ether bath.
  • n-BuLi 21 mmol, 1.6 M in cyclohexanes
  • the reaction mixture was allowed to warm to ⁇ 70° C. and then quenched by adding of 21 mL of 1 N HCl.
  • the reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organic layer was washed with saturated sodium chloride, then concentrated to a residue.
  • the residue was chromatographed on silica gel using 1:1 ethyl acetate:hexanes to give the product. Typical yields are 30-60%.
  • a substituted 2-aminobenzophenone (1 mmol) was dissolved in 5 mL of pyridine.
  • a substituted sulfonyl chloride (1.3 mmol) and dimethlaminopyridine (10 mg) were added to the 2-aminobenzophenone solution.
  • the reaction mixture was heated to 110° C. for 2 hours.
  • the solvent was evaporated under a stream of nitrogen and the product was isolated by reverse phase HPLC using the method given below. Typical yields are 60-80%.
  • Solvent A 99% Water/1% CH 3 CN/0.1% Formic Acid
  • Solvent B 95% CH 3 CN/5% Water/0.1% Formic Acid
  • the gradients were programmed according to their analytical scale HPLC retention time (t ana ) which was obtained by running a linear gradient of 0% B to 100% B in A in 3.5 min on a Phenomenex Luna 5 micron C18(2) 50 ⁇ 4.6 mm column at 3.5 mL/min.
  • FMAT Assay determine inhibitors of TECK binding to human CCR9 receptors:
  • a CCR9 assay buffer was prepared by adding 5 mL of 1M HEPES, 1.25 mL of 1M CaCl 2 , 1.5 mL of 1M MgCl 2 , 14 MgCl 2 , mL of 5M NaCl, 0.5 g fatty acid free bovine serum albumin, and 1 mL of 5% azide were added to 400 mL of distilled, deionized water (ddH 2 O). The solution was mixed until all the fatty acid free bovine serum albumin was dissolved. After pH was adjusted to 7.4, the final volume was adjusted to 500 mL, and the solution was filtered through a 0.2 ⁇ m filter.
  • a 1000 nM biotinylated human TECK in assay buffer was prepared.
  • a 14 ⁇ M solution of Cy5- ⁇ -Biotin working solution was prepared.
  • Biotinylated TECK and Cy5- ⁇ -Biotin working solutions were mixed right before cells were ready to be added into wells.
  • 13 mL of assay buffer was added to a 50 mL polyethylene tube.
  • 5.6 ⁇ L of 1000 nM biotinylated TECK working solution and 178 ⁇ L of 14 ⁇ g/mL Cy5- ⁇ -Biotin working solution were added to the tube and mixed.
  • the final biotinylated TECK concentration was 0.4 nM and the final Cy5- ⁇ -Biotin concentration 0.064 ⁇ g/mL.
  • Cells were plated right before all other reagents are added to wells. Cells were collected and centrifuged in a tabletop centrifuge at 1200 rpm for 5 minutes. The supernatant was removed by vacuum without disturbing cell pellet. The cells were resuspended in 5 mL of assay buffer. The cells were counted by adding 1 mL of a cell suspension to an eppendorf tube. Then 150 ⁇ L of this suspension was added to 150 ⁇ L of Trypan Blue. Then 10 ⁇ L of Trypan Blue suspension was added to hemocytometer, and the cell number per mL was calculated. The cells were resuspended with CCR9 assay buffer to final 0.2 ⁇ 10 6 cells/mL for the assay. Approximately 2.6 ⁇ 10 6 cells for 1 384 well plate was needed.
  • FLIPR Assay determines inhibitors of TECK induced response in Chinese Hamster Ovary (CHO) cells that over express recombinant human CCR9 and the Ga16 protein.
  • CHO cells were diluted to give appropriately 10,000 cells/well (in a volume of 50 mL). Each well of the 384 black/clear plate was then seeded with 50 mL of the diluted cell suspension. The cell plates were placed in a 37° C. tissue culture incubator at 6% CO 2 overnight.
  • a wash buffer and a dye loading solution were prepared on the day the assay was preformed.
  • the wash buffer was prepared by mixing 880 mL of Nanopure water, 100 mL of 10 ⁇ HBSS and 20 mL of 1M HEPES to give 1 L of a 1 ⁇ HBSS and 20 mM HEPES solution.
  • 1 g of BSA (bovine serum albumin) was added to the 1 ⁇ HBSS/20 mM HEPES solution.
  • a 250 mM probenecid stock solution was prepared by dissolving 710 mg of probenecid in 5 mL of 1 N NaOH and 5 mL of previously prepared 1 ⁇ HBSS/20 mM HEPES/0.1% BSA buffer.
  • the 2 ⁇ FLUO-3 dye loading solution prepared by adding 22 mL of DMSO (100%) to each of 2 ⁇ 50 mg vials of FLUO-3. The vials were vortexed, then 22 mL of 20% pluronic acid was added to each 50 mg vial of FLUO-3 and vortex. 88 mL of reconstituted FLUO-3 stock solution was added to 11 mL of 1 ⁇ wash buffer.
  • Each cell plate was washed with the wash buffer. At the end of the wash, there was 25 mL residual volume per well. 25 mL of dye loading solution was added to each well of the 384 plate. The plates were placed in an incubator for at least 1 hour. A yellowplate calibration plate was run on a FLIPR 384, and a standard deviation of less than 3.5% was obtained.
  • Ligand (rhTECK) plate at 4 ⁇ ligand EC 50 (final assay concentration) in wash buffer was prepared and plated into a Greiner 384 well plate. Appropriate amount of 4 ⁇ ligand was added to wells A1 through P10 and A13 through P22. In columns 11 and 12, FLIPR wash buffer was added to wells A11-D12 and 4 ⁇ ligand solution was added to wells E11-H12.
  • the cell plates were incubated at least a 1 hour with the dye solution, then washed with wash buffer. At the end of the wash, there was 25 mL residual volume per well.
  • the cell plate were loaded onto the stacker of FLIPR-384.
  • Individual well activity (IWA) was measured, using the max-min function between timepoints 88-145. Data was expressed as % inhibition of rhTECK induced response, and IC 50 values were calculated for compounds displaying antagonist activity.
  • IC 50 value less than or equal to about 1.0 ⁇ M: 17-21, 31-34, 44, 70, 73, 74, and 115.
  • IC 50 value less than or equal to about 1.0 ⁇ M: 8-13, 14-15, 17-20, 22-24, 31-36, 44, 49, 63, and 69-89, 116-143.

Abstract

The invention relates to compounds represented by Structural Formula I, which can bind to CCR9 receptors and block the binding of a ligand (e.g., TECK) to the receptors. The invention also relates to a method of inhibiting a function of CCR9, and to the use compounds represented by Structural Formula I in research, therapeutic, prophylactic and diagnostic methods.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a continuation of U.S. patent application Ser. No. 12/640,439, filed Dec. 17, 2009 (pending), which is a continuation of U.S. patent application Ser. No. 11/974,850, filed Oct. 16, 2007, now U.S. Pat. No. 7,659,272, which is a divisional of U.S. patent application Ser. No. 11/391,633, filed Mar. 28, 2006, now U.S. Pat. No. 7,282,502, which is a continuation of U.S. patent application Ser. No. 10/443,155, filed May 21, 2003, now U.S. Pat. No. 7,238,717, which claims the benefit of U.S. Provisional Application Ser. No. 60/383,573, filed May 24, 2002 (abandoned). The entire contents of each of the above-referenced patent applications are incorporated herein by this reference.
  • BACKGROUND OF THE INVENTION
  • Chemokines are a large and growing family of nearly forty 6-14 kD (non-glycosylated) heparin binding proteins that mediate a wide range of biological functions (Taub, D. D. and Openheim, J. J., Ther. Immunol., 1:229-246 (1994)). The chemokines can be divided into families based on the position of four cysteine residues that form two disulfide bonds (Kelner, G. S., et al., Science, 266:12395-1399 (1994); Bazan, J. F., et al., Nature, 385:640-644 (1997); Pin, Y., et al., Nature, 385:611-617 (1997)). Chemokine receptors can also be divided into families based on the type of chemokine they bind, although, no clear structural differences have been identified that distinguish the receptor sub-families (Mackay, C. R., J. Exp. Med., 184:799-802 (1996)).
  • Chemokines play a vital role in leukocyte adhesion and extravasation. For example, in various in vitro assays, chemokines can induce the chemotaxis or transendothelial migration of leukocytes (Taub, D. D. and Openheim, J. J., Ther. Immunol., 1:229-246 (1994)), while in vivo injection (Taub, D. D., et al., J. Clin. Invest., 97:1931-1941 (1996)) or over-expression of chemokines (Fuentes, M. E., et al., J. Immunol., 155:5769-5776 (1995)) can result in leukocyte accumulation at the site of chemokine injection or expression. Antagonists of chemokines can prevent leukocyte trafficking (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993)) and may have beneficial effects on acute and chronic inflammation (Sekido, N., et al., Nature, 365:654-657 (1993); Karpus, W. J., et al., J. Immunol., 155:5003-5010 (1995)). Chemokines have also been reported to modulate angiogenesis (Gupta, S. K., et al., Proc. Natl. Acad. Sci. USA, 92:7799-7803 (1995)), hematopoiesis (Taub, D. D. and Openheim, J. J., Ther. Immunol., 1:229-246 (1994)) as well as T lymphocyte activation (Zhou, Z., et al., J. Immunol.
  • 151:4333-4341 (1993); Taub, D. D., et al., J. Immunol., 156:2095-2103 (1996)). In addition, several chemokine receptors act as co-receptors, along with CD4, for entry of M tropic and T tropic HIV-1 (Choe, H., et al., Cell, 85:1135-1148 (1996); Feng, Y., et al., Science, 272:872-877 (1996)).
  • Several subsets of CD4 lymphocytes can be defined based on their expression of various adhesion molecules that are known to effect trafficking to different physiologic sites (Mackay, C. R., Curr. Opin. Immunol., 5:423-427 (1993)). For example, CLA+ve memory CD4 lymphocytes traffic to the skin (Berg, E. L., et al., Nature, 174(6):1461-1466 (1991)), while CLA−ve α4β7+ve memory CD4 lymphocytes traffic to mucosal sites (Hamman, A., et al., J. Immunol., 152:3282-3292 (1994)). Leukocyte adhesion to endothelium is thought to involve several overlapping steps including rolling, activation and arrest. Rolling leukocytes are exposed to factors expressed at the adhesion site resulting in activation of the leukocyte and up-regulation of integrin-mediated adhesion. As a consequence of such integrin-mediated interactions, leukocytes arrest on the endothelium (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993); Bargatze, R. F., et al., Immunity, 3:99-108 (1995)). Leukocyte activation and up-regulation of integrin molecules occurs via a pertussis toxin sensitive mechanism that is thought to involve chemokine receptors (Bargatze, R. F. and Butcher, E. C., J. Exp. Med., 178:367-372 (1993); Campbell, J. J., et al., Science, 279:381-383 (1998)).
  • Memory CD4+ lymphocytes can be grouped based upon the expression of certain chemokine receptors. For example, CXCR3, CCR2 and CCR5 (Qin, S., et al., Eur. J. Immunol., 26:640-647 (1996); Qin, S., et al., J. Clin. Invest., 101:746-754 (1998); Liao, F., et al., J. Immunol., 162:186-194 (1999)) are all expressed on subsets of memory CD4 lymphocytes, and certain chemokines act selectively on naive T cells (Adema, G. J., et al., Nature, 387:713-717 (1997)). Furthermore, several chemokines which are ligands for such receptors have been shown to be expressed in inflammatory sites (Gonzalo, J. A., et al., J. Clin. Invest., 98:2332-2345 (1996)) and in some cases in lymph nodes draining a challenged site (Tedla, N., et al., J. Immunol., 161:5663-5672 (1998)). In vitro derived TH1/TH2 lymphocyte lines have also been shown to differentially express chemokine receptors. Specifically, TH1 lymphocytes have been shown to selectively express CXCR3 and CCR5, while TH2 lymphocytes selectively express CCR4, CCR8 and CCR3 (Bonecchi, R. G., et al., J. Exp. Med., 187:129-134 (1998); Sallusto, F. D., et al., J. Exp. Med., 187:875-883 (1998); Sallusto, F., Science, 277:2005-2007 (1997); Andrew, D. P., et al., J. Immunol. 161:5027-5038 (1998); Zingoni, A., et al., J. Immunol., 161:547-555 (1998)). Interestingly, in some cases the chemokines for these respective chemokine receptors, such as MDC for CCR4 and IP-10 for CXCR3, are induced by cytokines associated with a TH1/TH2 environment (Andrew, D. P., et al., J. Immunol. 161:5027-5038 (1998); Luster, A. D., et al., Nature, 315:672-676 (1985)).
  • SUMMARY OF THE INVENTION
  • The present invention relates to compounds represented by Structural Formula I, and pharmaceutically acceptable salts, solvates, and hydrates of such compounds:
  • Figure US20100324093A1-20101223-C00001
  • In Structural Formula I, Y is C(O), O, S, S(O) or S(O)2; X1, X2, and X3 are each, independently, N or CR, provided that at least one of X1, X2, or X3 is CR; R, for each occurrence, and R1 are each, independently, H or a substituent. In preferred embodiments, the substituents at positions R and R1 are each, independently, an aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, sulonamido, sulfone, sulfoxide, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12. R11 and R12 are defined further herein; R6 is H, an aliphatic carbonyl group, or an aliphatic ester; and Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group. Preferably, Ar1 is a substituted or unsubstituted phenyl, a substituted or unsubstituted pyridyl, a substituted or unsubstituted pyrimidinyl, or a substituted or unsubstituted pyrazinyl. Preferably, Ar2 is a substituted or unsubstituted phenyl, or a substituted or unsubstituted pyridyl. More preferably, Ar1 is a substituted phenyl and Ar2 is substituted pyridyl. Ring A is substituted or unsubstituted.
  • In a preferred embodiment, compounds of the invention are represented by Structural Formula II:
  • Figure US20100324093A1-20101223-C00002
  • In Structural Formula II, X4 is N, N+—O, or CR; and ring A, ring B, and ring C are each, independently, substituted or unsubstituted. R is H or a substituent. Examples of substituents for R are defined as in Structural Formula I.
  • In a more preferred embodiment, compounds of the invention are represented by Structural Formula III:
  • Figure US20100324093A1-20101223-C00003
  • In Structural Formula III, X4 is N, N+—O or CR; ring A, ring B, and ring C are each, independently, substituted or unsubstituted, and R is H or a substituent. Examples of substituents for R are defined as in Structural Formula I; and R8 is H or an electron withdrawing group. Preferably, R8 is a halo, nitro, alkylcarbonyl or trihaloalkyl. More preferably, R8 is Cl, Br or NO2.
  • In another preferred embodiment, compounds of the invention are represented by Structural Formula IV.
  • Figure US20100324093A1-20101223-C00004
  • In Structural Formula IV, Ar1, Ar2, R1 and R6 are defined as in Structural Formula I; and R19 and R20 are each, independently, H or a substituent. Examples of substituents in the R19 and R20 positions include an aliphatic group, a haloalkyl group, an ester, an amide, alkylcarbonyl, a halogen, COOH, NO2, alkoxy, haloalkoxy, CN, amino, and aminoalkyl.
  • In another preferred embodiment, compounds of the invention are represented by Structural Formula V.
  • Figure US20100324093A1-20101223-C00005
  • In Structural Formula V, X4 is CR, N or N+—O; X1, X2, X3, R, and Ar2 are defined as in Structural Formula I; R1, R2, R3, R4, R5, are, independently, H, an aliphatic group, a haloalkyl group, a halo, COOH, NO2, or an alkoxy, a haloalkoxy.
  • In a more preferred embodiment, compounds of the invention are represented by Structural Formula VI.
  • Figure US20100324093A1-20101223-C00006
  • In Structural Formula VI, X1 is defined as in Structural Formula I; X4 is defined as in Structural Formula V; R8 is H or an electron withdrawing group; m and n are each, independently, 0 or an integer from 1 to 3; each R9 is, independently, aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12; or two adjacent R9 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1, or 2 heteroatoms selected from N, O, and S; each R10 is, independently, halo, aliphatic group, alkoxy, or haloalkyl; or two adjacent R10 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from N, O, and S; each R11 is, independently, selected from H or an aliphatic group; and R12 is an aliphatic group.
  • In another preferred embodiment, compounds of the invention are represented by Structural Formula VII.
  • Figure US20100324093A1-20101223-C00007
  • In Structural Formula VII, ring B is substituted or unsubstituted; X1 is defined as in Structural Formula I; R8 is defined as in Structural Formula VI; p is 0 or an integer from 1-3; and each R13 is, independently, a halo or a substituted or unsubstituted heteroaryl.
  • In another preferred embodiment, compounds of the invention are represented by Structural Formula VIII.
  • Figure US20100324093A1-20101223-C00008
  • In Structural Formula VIII, ring A, Ar1, Ar2, X1, R1, R6 are defined as in Structural Formula I; R19 and R20 are defined as in Structural Formula IV; and Y1 is S, O, S(O), or S(O)2.
  • The present invention provides a method of inhibiting a CCR9 receptor by contacting the receptor with an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof.
  • In one embodiment, the invention provides a method of inhibiting CCR9-mediated homing of leukocytes in a subject by administering to the subject an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof. In a preferred embodiment, the method inhibits homing of leukocytes to mucosal tissue.
  • In another embodiment, the invention provides a method of treating a subject having a disease that is mediated by a CCR9 receptor, such as an inflammatory disease, for example Celiac's disease or an inflammatory bowel disease, by administering to the subject an effective amount of a compound of the present invention, and/or pharmaceutically acceptable salts, solvates and hydrates thereof.
  • The present invention provides a pharmaceutical composition having a pharmaceutically acceptable carrier and at least one compound of the present invention.
  • Chemokines and their associated receptors (e.g., TECK and CCR9, respectively) are proinflammatory mediators that promote recruitment and activation of multiple lineages of leukocytes and lymphocytes. Continuous release of chemokines at sites of inflammation mediates the ongoing migration of effector cells in chronic inflammation. CCR9 and its associated chemokine TECK, have been implicated in chronic inflammatory diseases, such as inflammatory bowel diseases. Small molecule inhibitors of the interaction between CCR9 and its ligands (e.g., TECK), such as the compounds of the invention, are useful for inhibiting harmful inflammatory processes triggered by receptor-ligand interactions and thus are useful for treating diseases mediated by CCR9, such as chronic inflammatory diseases.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The term “aliphatic” as used herein means straight-chain, branched or cyclic C1-C12 hydrocarbons which are completely saturated or which contain one or more units of unsaturation but which are not aromatic. For example, suitable aliphatic groups include substituted or unsubstituted linear, branched or cyclic alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. The terms “alkyl”, “alkoxy”, and “alkylthio”, used alone or as part of a larger moiety includes both straight and branched chains containing one to twelve carbon atoms. The terms “alkenyl” and alkynyl” used alone or as part of a larger moiety shall include both straight and branched chains containing two to twelve carbon atoms. The term “cycloalkyl” used alone or as part of a larger moiety shall include cyclic C3-C12 hydrocarbons which are completely saturated or which contain one or more units of unsaturation, but which are not aromatic.
  • As used herein, aryl groups are carbocyclic aromatic ring systems (e.g. phenyl), fused polycyclic aromatic ring systems (e.g. naphthyl and anthracenyl) and aromatic ring systems fused to carbocyclic non-aromatic ring systems (e.g., 1,2,3,4-tetrahydronaphthyl) having six to about fourteen carbon atoms.
  • The terms “haloalkyl”, “haloalkenyl” and “haloalkoxy” means alkyl, alkenyl or alkoxy, as the case may be, substituted with one or more halogen atoms. The term “halogen” means F, Cl, Br or I.
  • The term “aryl” used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to aromatic ring groups having five to fourteen members, such as phenyl, benzyl, phenethyl, 1-napthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl. The term “aryl” also refers to rings that are optionally substituted. The term “aryl” may be used interchangeably with the term “aryl ring”. “Aryl” also includes fused polycyclic aromatic ring systems in which an aromatic ring is fused to one or more rings. Examples include 1-naphthyl, 2-naphthyl, 1-anthracyl and 2-anthracyl. Also included within the scope of the term “aryl”, as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as in an indanyl, phenantriidinyl, or tetrahydronaphthyl, where the radical or point of attachment is on the aromatic ring.
  • The term “heteroatom” means nitrogen, oxygen, or sulfur and includes any oxidized form of nitrogen and sulfur, and the quaternized form of any basic nitrogen. Also the term “nitrogen” includes a substitutable nitrogen of a heterocyclic ring. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR18 (as in N-substituted pyrrolidinyl).
  • The term “heterocycle”, as used herein includes non-aromatic ring systems having five to fourteen members, preferably five to ten, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, or S. Examples of heterocyclic rings include 3-1H-benzimidazol-2-one, 3-tetrahydrofuranyl, 2-tetrahydropyranyl, 3-tetrahydropyranyl, 4-tetrahydropyranyl, [1,3]-dioxalanyl, [1,3]-dithiolanyl, [1,3]-dioxanyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3-morpholinyl, 4-morpholinyl, 2-thiomorpholinyl, 3-thiomorpholinyl, 4-thiomorpholinyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrorolidinyl, 1-piperazinyl, 2-piperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, N-substituted diazolonyl, 1-pthalimidinyl, benzoxanyl, benzopyrrolidinyl, benzopiperidinyl, benzoxolanyl, benzothiolanyl, and benzothianyl. Also included within the scope of the term “heterocycle”, as it is used herein, is a group in which a non-aromatic heteroatom-containing ring is fused to one or more aromatic or non-aromatic rings, such as in an indolinyl, chromanyl, phenantrhidinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the non-aromatic heteroatom-containing ring. The term “heterocycle”, whether saturated or partially unsaturated, also refers to rings that are optionally substituted.
  • The term “heteroaryl”, used alone or as part of a larger moiety as in “heteroaralkyl” or “heteroarylalkoxy”, refers to heteroaromatic ring groups having five to fourteen members. Examples of heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxadiazolyl, 5-oxadiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 3-pyridazinyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-triazolyl, 5-triazolyl, 2-thienyl, 3-thienyl, thianaphthenyl, carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, benzotriazolyl, benzothiazolyl, benzooxazolyl, benzimidazolyl, isoquinolinyl, indolyl, isoindolyl, acridinyl, or benzoisazolyl. Also included within the scope of the term “heteroaryl”, as it is used herein, is a group in which a heteroatomic ring is fused to one or more aromatic or nonaromatic rings where the radical or point of attachment is on the heteroaromatic ring. Examples include tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[3,4-d]pyrimidinyl. The term “heteroaryl” also refers to rings that are optionally substituted. Preferred heteroaryl groups are thienyl, benzo(b)thienyl, pyrrolyl, indolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, furanyl, and benzo(b)furanyl. More preferred heteroaryl groups are pyridyl and thienyl.
  • An arylalkyl group, as used herein, is an aryl substituent that is linked to a compound by an alkyl group having from one to twelve carbon atoms.
  • An alkoxy group, as used herein, is a C1-C12 alkyl group that is connected to a compound via an oxygen atom. Examples of alkoxy groups include but are not limited to methoxy, ethoxy, propoxy, isopropoxy, and t-butoxy.
  • A cycloalkoxy group, as used herein, is a cyclic C3-C12 hydrocarbon which is attached to a compound via an oxygen. Cycloalkoxy groups include but are not limited to cyclopropoxy and cyclobutoxy.
  • A haloalkoxy, as used herein, is a haloalkyl group that is attached to a compound via an oxygen. A preferred haloalkoxy is trifluoromethoxy.
  • An aryloxy, as used herein, is an aryl group that is attached to a compound via an oxygen. A preferred aryloxy is phenoxy.
  • A arylalkoxy group, as used herein, is a arylalkyl group that is attached to a compound via an oxygen on the C1-C12 alkyl portion of the arylalkyl. A preferred arylalkoxy is phenylmethoxy.
  • An alklythio group, as used herein, is a C1-C12 alkyl group that is connected to a compound via a sulfur atom.
  • A aliphatic carbonyl group, as used herein, is an aliphatic group that is connected to a compound via a carbonyl group. A preferred aliphatic carbonyl is acetyl.
  • A aliphatic ester group, as used herein, an aliphatic group that is connected to a compound via an ester linkage (i.e., —C(O)O-aliphatic group).
  • An electron withdrawing group is a group which causes a dipole moment in the direction of the group. Suitable electron withdrawing groups include but are not limited to halo (preferably chloro), haloalkyl (preferably trifluoromethyl), nitro, cyano, sulfonamido, sulfone, and sulfoxide.
  • An aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) may contain one or more substituents. Examples of suitable substituents include aliphatic groups, aryl groups, haloalkoxy groups, heteroaryl groups, halo, hydroxy, OR14, COR14, COOR14, NHCOR14, OCOR14, benzyl, haloalkyl (e.g., trifluoromethyl and trichloromethyl), cyano, nitro, S(O), S(O)2, SO3 , SH, SR14, NH2, NHR14, NR14R15, NR6S(O)2—R7, and COOH, wherein R14 and R15 are each, independently, an aliphatic group, a cycloalkyl, an aryl group, or an arylalky group. Other substituents for an aryl or heteroaryl group include —R16, —OR16, —SR16, 1,2-methylene-dioxy, 1,2-ethylenedioxy, protected OH (such as acyloxy), phenyl (Ph), substituted Ph, —O(Ph), substituted —O(Ph), —CH2(Ph), substituted —CH2CH2(Ph), substituted —CH2CH2(Ph), —N(R16)2, —NR16CO2R16, —NR16NR16C(O)R16, —NR16R16C(O)N(R16)2, —NR16NR16CO2R16, —C(O)C(O)R16, —C(O)CH2C(O)R16, —CO2R16, —C(O)R16, —C(O)N(R16)2, —OC(O)N(R16)2, —S(O)2R16, —SO2N(R16)2, —S(O)R16, —NR16SO2N(R16)2, —NR16SO2R16, —C(═S)N(R16)2, —C(═NH)—N(R16)2, —(CH2)yNHC(O)R16, —(CH2)yNHC(O)CH(V—R16)(R16); wherein R16 is hydrogen, a substituted or unsubstituted heteroaryl or heterocyclic ring, phenyl (Ph), substituted Ph, —O(Ph), substituted —O(Ph), —CH2 (Ph), or substituted —CH2 (Ph); y is 0-6; and V is a linker group. Preferred substituents on the aryl or heteroaryl group include halo, haloalkyl, sulfone, sulfoxide, nitro, cyano, alkyl group, alkoxy group, and alkylamino group. Examples of substituents on the aliphatic group or the phenyl ring include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl. Substituents for an aryl or a heteroaryl group include aryl groups (including a carbocyclic aryl group or a heteroaryl group), aliphatic groups, cycloalkyl groups, haloalkoxy groups, heteroaryl groups, hydroxy, OR14, aldehyde, COR14, COOR14, NHCOR14, OCOR14, benzyl, haloalkyl (e.g., trifluoromethyl and trichloromethyl), halo, cyano, nitro, SO3 , SH, SR14, NH2, NHR14, NR14R15, NR6S(O)2—R7, or COOH, wherein R14 and R15 are each, independently, an aliphatic group, a cycloalkyl, an aryl group, or an arylalky group.
  • An aliphatic group or a heterocycle may contain one or more substituents. Examples of suitable substituents on the saturated carbon of an aliphatic group of a heterocycle include those listed above for an aryl or heteroaryl group and the following: ═O, ═S, ═NNHR17, ═NN(R17)2, ═NNHC(O)R17, ═NNHCO2(alkyl), ═NNHSO2(alkyl), or ═NR17, where each R17 is independently selected from hydrogen, an unsubstituted aliphatic group or a substituted aliphatic group. Examples of substituents on the aliphatic group include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkoxy, thioalkyl, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • Suitable substitutents on the nitrogen of a non-aromatic heterocycle or on an unsaturated nitrogen of a heteroaryl include —R18, —N(R18)2, —C(O)R18, —CO2R18, —C(O)C(O)R18, —C(O)CH2C(O)R18, —SO2R18, —SO2N(R18)2, —C(═S)N(R18)2, —C(═NH)—N(R18)2, and —NR18SO2R18; wherein R18 is hydrogen, an aliphatic group, a substituted aliphatic group, phenyl (Ph), substituted Ph, —O(Ph), substituted —O(Ph), —CH2(Ph), or an unsubstituted heteroaryl or heterocyclic ring. Examples of substituents on the aliphatic group or the phenyl ring include amino, alkylamino, dialkylamino, aminocarbonyl, halogen, alkyl, alkylaminocarbonyl, dialkylaminocarbonyloxy, alkoxy, nitro, cyano, carboxy, alkoxycarbonyl, alkylcarbonyl, hydroxy, haloalkoxy, or haloalkyl.
  • The term “linker group” or “linker” means an organic moiety that connects two parts of a compound. Linkers are typically comprised of an atom, such as oxygen or sulfur, a unit, such as —NH—, —CH2—, —C(O)—, or —C(O)NH—, or a chain of atoms, such as an alkylene chain. The molecular mass of a linker is typically in the range of about 14 to 200, preferably in the range of 14 to 96 with a length of up to about six atoms. Examples of linkers include a saturated or unsaturated C1-C6 alkylene chain which is substituted or unsubstituted, and wherein one or two saturated carbons of the chain are optionally replaced by —C(O)—, —C(O)C(O)—, —CONH—, —CONHNH—, —CO2—, —OC(O)—, —NHCO2—, —O—, —NHCONH—, —OC(O)NH—, —NHNH—, —NHCO—, —S—, —SO—, —SO2—, —NH—, —SO2NH—, or —NHSO2—.
  • It will be apparent to one skilled in the art that certain compounds of this invention may exist in tautomeric forms, all such tautomeric forms of the compounds being within the scope of the invention. Unless otherwise stated, structures depicted herein are also meant to include all stereochemical forms of the structure; i.e., the R and S configurations for each asymmetric center. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.
  • The compounds of the present invention have IC50s of less than 1 μm; less than 750 nm; preferably less than 500 nm; more preferably less than 250 nm; even more preferably less than 100 nm, most preferably less than 50 nm; and the most preferable less than 10 nm, e.g., less than 5 nm.
  • As used herein “mammalian CCR9” refers to naturally occurring or endogenous mammalian CCR9 proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian CCR9 protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature receptor protein, polymorphic or allelic variants, and other isoforms of a mammalian CCR9 (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated, unglycosylated). Naturally occurring or endogenous mammalian CCR9 proteins (see e.g., GenBank Accession Numbers NM031200 and U45982 and Yu et al. (2000) J. Immunol. 164:1293-1305 which describe various forms of naturally occurring mammalian CCR9) include wild type proteins such as mature CCR9, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces mammalian CCR9, for example. Polymorphic, allelic, splice and other naturally occurring variants of mammalian CCR9 can be expressed in particular organs, tissues or cells and have altered properties (e.g., altered affinity for ligand (e.g. TECK)) and specialized biological function (e.g., T cell development, T cell recruitment). Naturally occurring or endogenous mammalian CCR9 proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding mammalian CCR9, are referred to by the name of the corresponding mammal. For example, where the corresponding mammal is a human, the protein is designated as a human CCR9 protein (e.g., a recombinant human CCR9 produced in a suitable host cell).
  • As used herein “mammalian TECK” refers to naturally occurring or endogenous mammalian TECK proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian TECK protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature receptor protein, polymorphic or allelic variants, and other isoforms of a mammalian TECK (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated, unglycosylated). Naturally occurring or endogenous mammalian TECK proteins (see e.g., GenBank Accession Number U86358 and Vicari et al. (1997) Immunity 7:291-301 which describe naturally occurring mammalian TECK) include wild type proteins such as mature TECK, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces mammalian TECK, for example.
  • The term “pharmaceutically acceptable” means that the carrier, diluent, excipients and salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof. Pharmaceutical formulations of the present invention are prepared by procedures known in the art using well known and readily available ingredients.
  • “Preventing” refers to reducing the likelihood that the recipient will incur or develop any of the pathological conditions described herein, for example inhibiting the onset of these symptoms.
  • “Treating” refers to eliminating or reducing the severity of a pathological condition described herein and/or eliminating, alleviating and/or reducing the symptoms of a pathological condition described herein.
  • Compounds of the invention can be used to treat a subject having an inflammatory disease. In one embodiment, the method is a method of inhibiting a CCR9 function in a subject. In particular embodiments, the method is a method of treating a subject having an inflammatory disease associated with mucosal tissue, such as Crohn's disease, colitis, or Celiac disease. In another embodiment, the method is a method of inhibiting CCR9-mediated homing of leukocytes in a subject. See Papadakis, et al., Gastroenterology (2001), 121:246-254 for a discussion of the relationship of CCR9 receptors to the above diseases or conditions.
  • In one embodiment, the compounds of the invention are represented by any one of Structural Formulas IX-XXIV:
  • Figure US20100324093A1-20101223-C00009
    Figure US20100324093A1-20101223-C00010
    Figure US20100324093A1-20101223-C00011
  • In Structural Formulas IX-XXV, Ar2 and Y are defined as in Structural Formula I; and rings A and B are substituted or unsubstituted.
  • In another embodiment of the invention, compounds used in the method of the invention and in pharmaceutical compositions of the invention are represented by Structural Formula XXVI or XXVII:
  • Figure US20100324093A1-20101223-C00012
  • In Structural Formulas XXVI and XXVII, R8 is defined as in Structural Formula V, and R13 and p are defined as in Structural Formula VII.
  • In a first preferred embodiment of the invention, Y in Structural Formula I, or IX-XXIV is C═O.
  • In a second preferred embodiment, ring C in Structural Formula II or III or Ar2 in Structural Formula I, IV, V or in any one of Structural Formulas IX-XXV is unsubstituted or substituted with one or more substituents selected from aliphatic group (including substituted aliphatic groups such as haloalkyl) aryl, arylalkyl, alkoxy (including cycloalkoxy and substituted alkoxy groups such as haloalkoxy), aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, S(O)-(aliphatic), S(O)2-(aliphatic), NR11S(O)2-(aliphatic), C(O)N(R11)2, C(O)R12, N(R11)2, NR11C(O)2R12 and NR11C(O)R12, wherein R11 for each occurrence is, independently, H or an aliphatic group, and R12 is an aliphatic group. More preferred substitutents for ring C or R2 are selected from an aliphatic group, an alkoxy, and a haloalkoxy.
  • In a third preferred embodiment, ring A in Structural Formula I or IV, or one or both rings A and B in Structural Formula II or III or in any one of Structural Formulas IX-XXV are, independently, substituted with a substituent selected from halo, aliphatic group, alkoxy, and haloalkyl.
  • In a fourth preferred embodiment, ring A in Structural Formula I, II, III, IV, V, VI, VII, VIII, or in any one of Structural Formulas IX-XXV is substituted with an electron withdrawing substituent para to the sulfonamide group.
  • In a fifth preferred embodiment of the invention, X3 in Structural Formula V is CR21, wherein R21 is an electron withdrawing group. More preferably, R21 is halo, nitro, aliphatic carbonyl, or trihalomethyl. Most preferably, R21 is Cl, Br, or nitro.
  • In a sixth preferred embodiment of the invention, Ar2 in Structural Formula I, IV, V, VIII or in any one of Structural Formulas IX-XXV is a substituted or unsubstituted phenyl, a substituted or unsubstituted naphthyl, a substituted or unsubstituted thienyl, or a substituted or unsubstituted thianaphthenyl. More preferably, Ar2 is a substituted or unsubstituted phenyl or a substituted or unsubstituted thienyl.
  • In a seventh preferred embodiment of the invention, R9 for each occurrence in Structural Formula VI is, independently, an aliphatic group, an alkoxy, or a haloalkoxy.
  • In an eighth preferred embodiment of the invention, R8 in Structural Formula I, VI, VII, VIII, XXV and XXVI is a halo, nitro, alkylcarbonyl or trihaloalkyl. More preferably, R8 is Cl, Br or NO2.
  • In an ninth preferred embodiment of the invention, Y and Ar2 in Structural Formula I and VIII or in any one of Structural Formulas IX-XXV are defined as in the first and the sixth preferred embodiments, respectively.
  • In a tenth preferred embodiment of the invention, Ar2 Structural Formula V is defined as in the sixth preferred embodiments, and X3 is defined as in the fifth preferred embodiment.
  • In an eleventh preferred embodiment, X4 in Structural Formulas V or VI is an nitrogen oxide (N+—O).
  • In a twelfth preferred embodiment, n is one in Structural Formula VI and R9 is para to the sulfonamide substituent. More preferably, R9 is as defined in seventh preferred embodiment.
  • In a thirteen preferred embodiment, m is one in Structural Formula VI and R10 is meta to the carbonyl substituent.
  • In a fourteenth preferred embodiment, R6 in Structural Formula I, IV, or VIII is H.
  • Specific examples of compounds used in the method of the invention and in pharmaceutical compositions of the invention include but are not limited to the compounds listed in Table 1. Pharmaceutically acceptable salts, solvates and hydrates of the compounds listed in Table 1 are also useful in the method of the invention and in pharmaceutical compositions of the invention.
  • TABLE 1
    Specific compounds of the invention
    Example Compound Name
    7 N-[4-Chloro-2-(2-chloro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide
    8 Thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    9 N-(2-Benzoy-4-chloro-phenyl)-4-iodo-benzenesulfonamide
    10 N-(2-Benzoyl-4-chloro-phenyl)-4-chloro-benzenesulfonamide
    11 N-(2-Benzoyl-4-chloro-phenyl)-4-tert-butyl-benzenesulfonamide
    12 N-(2-Benzoyl-4-chloro-phenyl)-4-propyl-benzenesulfonamide
    13 N-(2-Benzoyl-4-chloro-phenyl)-4-ethyl-benzenesulfonamide
    14 N-(2-Benzoyl-4-nitro-phenyl)-4-ethyl-benzenesulfonamide
    15 N-(2-Benzoyl-4-chloro-phenyl)-4-isopropyl-benzenesulfonamide
    16 N-(2-Benzoyl-4-bromo-phenyl)-4-ethyl-benzenesulfonamide
    17 N-(2-Benzoyl-4-bromo-phenyl)-4-methoxy-benzenesulfonamide
    18 N-(2-Benzoyl-4-bromo-phenyl)-4-isopropyl-benzenesulfonamide
    19 4-Ethyl-N-[4-nitro-2-(3-trifluoromethyl-benzoyl)-phenyl]-
    benzenesulfonamide
    20 N-[4-Chloro-2-(3-methyl-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide
    21 N-(2-Benzoyl-4-bromo-phenyl)-4-chloro-benzenesulfonamide
    22 N-(2-Benzoyl-4-chloro-phenyl)-4-ethoxy-benzenesulfonamide
    23 N-(2-Benzoyl-4-chloro-phenyl)-4-propoxy-benzenesulfonamide
    24 N-(2-Benzoyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide
    25 N-(2-Benzoyl-4-chloro-phenyl)-4-butoxy-benzenesulfonamide
    26 N-(2-Benzoyl-4-chloro-phenyl)-4-benzyloxy-benzenesulfonamide
    27 N-(2-Benzoyl-4-chloro-phenyl)-4-phenoxy-benzenesulfonamide
    31 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide
    32 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethoxy-benzenesulfonamide
    33 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropyl-
    benzenesulfonamide
    34 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isobutyl-benzenesulfonamide
    35 N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-ethoxy-
    benzenesulfonamide
    36 N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-isopropoxy-
    benzenesulfonamide
    45 N-(3-Benzoyl-5-trifluoromethyl-pyridin-2-yl)-4-isopropoxy-
    benzenesulfonamide
    46 N-(2-Benzoyl-4-nitro-phenyl)-4-chloro-benzenesulfonamide
    47 5-Benzoyl-6-(4-isopropoxy-benzenesulfonylamino)-nicotinic acid
    48 N-(2-Benzoyl-4-chloro-phenyl)-2-nitro-benzenesulfonamide
    49 N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide
    50 N-(5-Benzoyl-pyrimidin-4-yl)-4-isopropoxy-benzenesulfonamide
    51 N-(3-Benzoyl-5-trifluoromethoxy-pyridin-2-yl)-4-isopropoxy-
    benzenesulfonamide
    52 N-(5-Benzoyl-2-trifluoromethyl-pyrimidin-4-yl)-4-isopropoxy-
    benzenesulfonamide
    53 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropoxy-benzenesulfonamide
    54 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclobutyl-benzenesulfonamide
    55 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-tert-butoxy-benzenesulfonamide
    56 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopentyl-benzenesulfonamide
    57 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylsulfanyl-
    benzenesulfonamide
    58 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylamino-benzenesulfonamide
    59 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropyl-benzenesulfonamide
    60 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclohexyl-benzenesulfonamide
    61 N-(3-Benzenesulfinyl-5-chloro-pyridin-2-yl)-4-isopropoxy-
    benzenesulfonamide
    62 N-(3-Benzenesulfonyl-5-chloro-pyridin-2-yl)-4-isopropoxy-
    benzenesulfonamide
    63 N-[4-Chloro-2-(pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy-
    benzenesulfonamide
    64 N-[4-Chloro-2-(pyridazine-4-carbonyl)-phenyl]-4-isopropoxy-
    benzenesulfonamide
    65 N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-
    isopropoxy-benzenesulfonamide
    66 N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-
    isopropoxy-benzenesulfonamide
    67 N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-
    isopropoxy-benzenesulfonamide
    68 N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-
    isopropoxy-benzenesulfonamide
    69 N-(2-Benzoyl-4-nitro-phenyl)-4-isopropyl-benzenesulfonamide
    70 N-(2-Benzoyl-4-nitro-phenyl)-4-isopropoxy-benzenesulfonamide
    71 N-(2-Benzoyl-4-nitro-phenyl)-4-methoxy-benzenesulfonamide
    72 N-(2-Benzoyl-4-chloro-phenyl)-4-trifluoromethoxy-benzenesulfonamide
    73 5-Oxazol-5-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    74 N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide
    75 N-(2-Benzoyl-4-chloro-phenyl)-4-methoxy-benzenesulfonamide
    76 N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide
    77 N-(2-Benzoyl-4-chloro-phenyl)-4-ethylamino-benzenesulfonamide
    78 5-(1-Methyl-5-trifluoromethyl-1H-pyrazol-3-yl)-thiophene-2-sulfonic acid (2-
    benzoyl-4-chloro-phenyl)-amide
    79 N-(2-Benzoyl-4-iodo-phenyl)-4-isopropoxy-benzenesulfonamide
    80 N-[4-Chloro-2-(3-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide
    81 N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-isopropyl-benzenesulfonamide
    82 N-(2-Benzoyl-4-chloro-phenyl)-4-nitro-benzenesulfonamide
    83 N-(2-Benzoyl-4-chloro-phenyl)-4-diethylamino-benzenesulfonamide
    84 N-(2-Benzoyl-4-fluoro-phenyl)-4-methoxy-benzenesulfonamide
    85 4-Chloro-N-[4-chloro-2-(pyridine-3-carbonyl)-phenyl]-benzenesulfonamide
    86 N-(2-Benzoyl-4-bromo-phenyl)-4-methyl-benzenesulfonamide
    87 N-(2-Benzoyl-4-chloro-phenyl)-4-bromo-benzenesulfonamide
    88 N-[4-Chloro-2-(pyridine-3-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide
    89 N-(2-Benzoyl-4-chloro-phenyl)-4-dimethylamino-benzenesulfonamide
    90 N-(2-Benzoyl-4-chloro-phenyl)-4-fluoro-benzenesulfonamide
    91 4-Chloro-N-[4-chloro-2-(2-fluoro-benzoyl)-phenyl]-benzenesulfonamide
    92 N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide
    93 Thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    94 5-Bromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    95 5-Chloro-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    96 N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide
    97 N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide
    98 4-Chloro-N-[4-chloro-2-(pyridine-2-carbonyl)-phenyl]-benzenesulfonamide
    99 N-(2-Benzoyl-4-chloro-phenyl)-benzenesulfonamide
    100 N-[4-(2-Benzoyl-4-chloro-phenylsulfamoyl)-phenyl]-acetamide
    101 N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide
    102 N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methyl-benzenesulfonamide
    103 N-(2-Benzoyl-4-chloro-phenyl)-4-cyano-benzenesulfonamide
    104 4,5-Dibromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    105 N-[4-Chloro-2-(pyridine-3-sulfonyl)-phenyl]-4-nitro-benzenesulfonamide
    106 Naphthalene-2-sulfonic acid [4-chloro-2-(pyridine-3-carbonyl)-phenyl]-amide
    107 N-(2-Benzoyl-4-chloro-phenyl)-3-bromo-benzenesulfonamide
    108 N-(2-Benzoyl-4-chloro-phenyl)-4-methyl-3-nitro-benzenesulfonamide
    109 N-[4-Chloro-2-(2-methoxy-benzoyl)-phenyl]-4-nitro-benzenesulfonamide
    110 N-(2-Benzoyl-4-chloro-phenyl)-2-trifluoromethyl-benzenesulfonamide
    111 5-Chloro-3-methyl-benzo[b]thiophene-2-sulfonic acid (2-benzoyl-4-chloro-
    phenyl)-amide
    112 N-(2-Benzoyl-phenyl)-4-nitro-benzenesulfonamide
    113 5-Isoxazol-3-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    114 N-(2-Benzoyl-phenyl)-4-methyl-benzenesulfonamide
    115 Benzo[b]thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    116 N-(4-Chloro-2-phenylsulfanyl-phenyl)-4-isopropoxy-benzenesulfonamide
    117 N-(2-Benzenesulfonyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide
    118 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-
    benzenesulfonamide
    119 N-[4-Chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-4-isopropoxy-
    benzenesulfonamide
    120 4-Isopropoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]-
    benzenesulfonamide
    121 4-Ethoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]-
    benzenesulfonamide
    122 N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropyl-
    benzenesulfonamide
    123 N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-
    benzenesulfonamide
    124 N-[5-Chloro-3-(thiophene-2-carbonyl)-pyridin-2-yl]-4-isopropoxy-
    benzenesulfonamide
    125 5-Oxazol-5-yl-thiophene-2-sulfonic acid [5-chloro-3-(3-fluoro-benzoyl)-
    pyridin-2-yl]-amide
    126 N-[3-(Benzofuran-2-carbonyl)-5-chloro-pyridin-2-yl]-4-isopropoxy-
    benzenesulfonamide
    127 N-(2-Benzoyl-4-trifluoromethyl-phenyl)-4-isopropoxy-benzenesulfonamide
    128 N-(3-Benzoyl-pyridin-4-yl)-4-isopropoxy-benzenesulfonamide
    129 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropyl-benzenesulfonamide
    130 N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide
    131 5-Oxazol-5-yl-thiophene-2-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-
    amide
    132 N-(2-Benzoyl-4-chloro-phenyl)-4-isopropylamino-benzenesulfonamide
    133 N-(4-Benzoyl-pyridin-3-yl)-4-isopropyl-benzenesulfonamide
    134 N-(4-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide
    135 N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropyl-benzenesulfonamide
    136 N-(2-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide
    137 N-[3-(3-Fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide
    138 N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropoxy-benzenesulfonamide
    139 4-Isopropoxy-N-[3-(pyridine-4-carbonyl)-pyridin-4-yl]-benzenesulfonamide
    140 6-Isopropoxy-pyridine-3-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-
    amide
    141 6-Isopropoxy-pyridine-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
    142 N-[4-Chloro-2-(pyridin-2-ylsulfanyl)-phenyl]-4-isopropyl-
    benzenesulfonamide
    143 N-(2-Benzoyl-phenyl)-4-isopropoxy-benzenesulfonamide
  • The structural formulas of Examples 7-27, 31-36, and 116-119 of Table 1 are shown in the Examples section. Examples 45-143 (Examples 116-119 are also included below) of Table 1 have the following structural formulas:
  • Figure US20100324093A1-20101223-C00013
    Figure US20100324093A1-20101223-C00014
    Figure US20100324093A1-20101223-C00015
    Figure US20100324093A1-20101223-C00016
    Figure US20100324093A1-20101223-C00017
    Figure US20100324093A1-20101223-C00018
    Figure US20100324093A1-20101223-C00019
    Figure US20100324093A1-20101223-C00020
    Figure US20100324093A1-20101223-C00021
    Figure US20100324093A1-20101223-C00022
    Figure US20100324093A1-20101223-C00023
    Figure US20100324093A1-20101223-C00024
    Figure US20100324093A1-20101223-C00025
    Figure US20100324093A1-20101223-C00026
    Figure US20100324093A1-20101223-C00027
    Figure US20100324093A1-20101223-C00028
    Figure US20100324093A1-20101223-C00029
    Figure US20100324093A1-20101223-C00030
    Figure US20100324093A1-20101223-C00031
  • The following compounds are preferred compounds for use in the method of the invention:
    • N-(2-Benzoyl-4-chlorophenyl)-4-ethyl-benzenesulfonamide;
    • N-(2-Benzoyl-4-nitrophenyl)-4-ethyl-benzenesulfonamide;
    • N-(2-Benzoyl-4-bromophenyl)-4-isopropyl-benzenesulfonamide;
    • N-(2-Benzoyl-4-chlorophenyl)-4-ethoxy-benzenesulfonamide;
    • N-(2-Benzoyl-4-chlorophenyl)-4-isopropoxy-benzenesulfonamide;
    • N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide;
    • N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethoxy-benzenesulfonamide;
    • N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropyl-benzenesulfonamide;
    • N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isobutyl-benzenesulfonamide;
    • N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide;
    • N-[4-Chloro-2-(pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide;
    • N-(2-Benzoyl-4-nitrophenyl)-4-isopropyl-benzenesulfonamide;
    • N-(2-Benzoyl-4-nitrophenyl)-4-isopropoxy-benzenesulfonamide;
    • N-(4-Chloro-2-phenylsulfanyl-phenyl)-4-isopropoxy-benzenesulfonamide;
    • N-(2-Benzenesulfonyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide;
    • N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide;
    • N-[4-Chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide;
    • 4-Ethoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]-benzenesulfonamide;
    • N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide;
    • N-(2-Benzoyl-4-trifluoromethyl-phenyl)-4-isopropoxy-benzenesulfonamide;
    • N-(3-Benzoyl-pyridin-4-yl)-4-isopropoxy-benzenesulfonamide;
    • N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropyl-benzenesulfonamide;
    • 5-Oxazol-5-yl-thiophene-2-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide;
    • 6-Isopropoxy-pyridine-3-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide;
    • 6-Isopropoxy-pyridine-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide;
    • N-[4-Chloro-2-(pyridin-2-ylsulfanyl)-phenyl]-4-isopropyl-benzenesulfonamide; and pharmaceutically acceptable salts, solvates, or hydrates thereof.
    Models of Inflammation
  • In vivo models of inflammation are available which can be used to assess the efficacy of compounds of the invention. For example, leukocyte infiltration upon intradermal injection of a chemokine and compound of the invention into a suitable animal, such as rabbit, mouse, rat, guinea pig or primate (e.g., rhesus macaque) can be monitored (see e.g., Van i, J. et al., J. Exp. Med., 176: 59-65 (1992); Zachariae, C. O. C. et al., J. Exp. Med. 171: 2177-2182 (1990); Jose, P. J. et al., J. Exp. Med. 179: 881-887 (1994)). In one embodiment, skin biopsies are assessed histologically for infiltration of leukocytes (e.g., CCR9+T cells). In another embodiment, labeled cells (e.g., stably transfected cells expressing a mammalian CCR9, labeled with 111In for example) capable of chemotaxis and extravasation are administered to the animal. For example, compound of the invention to be assessed which binds a mammalian CCR9 can be administered, either before, simultaneously with or after a CCR9 ligand or agonist (e.g., TECK) is administered to the test animal. A decrease of the extent of infiltration in the presence of the compound of the invention as compared with the extent of infiltration in the absence of said compound is indicative of inhibition.
  • As described herein, CCR9 is selectively expressed on memory lymphocytes which home to mucosal sites (e.g., CLA−ve α4β7hi CD4+ lymphocytes). Thus, animal models of inflammatory diseases of the mucosa (e.g., respiratory tract, urogenital tract, alimentary canal and associated organs and tissues (e.g., pancreas, liver, gall bladder)) can be used to assess the therapeutic efficacy of CCR9 inhibiting compounds. For example, the therapeutic efficacy of a compound of the invention can be studied in the cotton-top tamarin model of inflammatory bowel disease (Podolsky, D. K., et al., J. Clin. Invest. 92:372-380 (1993)). The CD45RBHi/SCID model provides a mouse model with similarity to both Crohn's disease and ulcerative colitis (Powrie, F. et al., Immunity, 1: 553-562 (1994)). Therapeutic efficacy in this model can be assessed, for example, by using parameters such as inhibition of recruitment of 111In-labeled cells to the colon and reduction in the number of CD4+T lymphocytes in the lamina propria of the large intestine after administration (e.g., intravenous (i.v.), intraperitoneally (i.p.) and per oral (p.o.)) of a compound.
  • Methods of Therapy
  • Inhibition of at least one function characteristic of a mammalian CCR9 protein according to the present invention provides an effective and selective way of inhibiting receptor-mediated functions. Once lymphocytes are recruited to a site, other leukocyte types, such as monocytes, may be recruited by secondary signals. Thus, compounds which can inhibit CCR9 function (e.g., compounds of the invention) can be used to inhibit leukocyte function (e.g., leukocyte infiltration including recruitment and/or accumulation).
  • In one aspect, the present invention provides a method of inhibiting an inflammatory response in a subject, comprising administering an effective amount of a compound of the invention which inhibits mammalian CCR9 function to a subject in need of such therapy. In one embodiment, an effective amount of a compound which inhibits one or more functions of a mammalian CCR9 protein (e.g., a human CCR9) is administered to a subject to inhibit (i.e., reduce or prevent) inflammation. Preferred compounds of the invention, inhibit an inflammatory response in a subject by inhibiting (i.e., reduce or prevent) binding of ligand (e.g. TECK) to CCR9. As a result, one or more inflammatory processes, such as leukocyte emigration, chemotaxis, exocytosis (e.g., of enzymes) or inflammatory mediator release, is inhibited. For example, leukocytic infiltration of inflammatory sites (e.g., in a inflamed mucus membrane (e.g., colon, small intestine)) can be inhibited according to the present method. In another embodiment, an effective amount of a compound of the invention which inhibits one or more functions of a mammalian CCR9 protein (e.g., a human CCR9) is administered to a subject to inhibit (i.e., reduce or prevent) CCR9-mediated homing of leukocytes. In particular embodiments, an effective amount of a compound which binds to human CCR9 and/or an effective amount of a compound which binds to human TECK is administered to a subject in need thereof.
  • Thus, the invention relates to a method of treating a subject having an inflammatory disease, comprising administering an effective amount of a compound of the invention that antagonizes CCR9 function. In a particular embodiment, the subject has an inflammatory bowel disease, such as Crohn's disease or colitis. Treatment includes therapeutic or prophylactic treatment. Treatment, in accordance with the method, can prevent disease or reduce the severity of disease in whole or in part.
  • The invention also relates to a method of inhibiting CCR9-mediated homing of leukocytes in a subject, comprising administering an effective amount of a compound of the invention that antagonizes CCR9 function, for example, the homing of leukocytes to mucosal sites can be inhibited. Immigration of circulating leukocytes into organs or tissue (e.g., intestine) and/or local recruitment of lymphocytes within an organ or tissue (e.g., IEL, LPL) can be inhibited in accordance with the method.
  • The term “subject” is defined herein to include animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent or murine species. A “subject in need of treatment to inhibit CCR9 function” is a subject in whom a beneficial therapeutic or prophylactic effect can be achieved by inhibiting CCR9 function. Examples include subjects with one of the diseases or conditions described herein.
  • Diseases and conditions associated with inflammation and/or infection can be treated using the methods described herein. In a preferred embodiment, the disease or condition is one in which the actions of lymphocytes, particularly lymphocytes which home to mucosal tissues, are to be inhibited or promoted for therapeutic (including prophylactic) purposes. In a particularly preferred embodiment, the inflammatory disease or condition is a T cell-mediated disease or condition.
  • Examples of inflammatory diseases associated with mucosal tissues which can be treated according to the present method include mastitis (mammary gland), vaginitis, cholecystitis, cholangitis or pericholangitis (bile duct and surrounding tissue of the liver), chronic bronchitis, chronic sinusitis, asthma, and graft versus host disease (e.g., in the gastrointestinal tract). As seen in Crohn's disease, inflammation often extends beyond the mucosal surface, accordingly chronic inflammatory diseases of the lung which result in interstitial fibrosis, such as interstitial lung diseases (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with rheumatoid arthritis, or other autoimmune conditions), hypersensitivity pneumonitis, collagen diseases, sarcoidosis, and other idiopathic conditions can be amenable to treatment. Pancreatitis and insulin-dependent diabetes mellitus are other diseases which can be treated using the present method.
  • In a particularly preferred embodiment, diseases which can be treated accordingly include inflammatory bowel disease (IBD), such as ulcerative colitis, Crohn's disease, ileitis, Celiac disease, nontropical Sprue, enteritis, enteropathy associated with seronegative arthropathies, microscopic or collagenous colitis, eosinophilic gastroenteritis, or pouchitis resulting after proctocolectomy, and ileoanal anastomosis.
  • Additional diseases or conditions, including chronic diseases, of humans or other species which can be treated with compounds of the invention that inhibit of CCR9 function, include, but are not limited to:
      • inflammatory or allergic diseases and conditions, including systemic anaphylaxis or hypersensitivity responses, drug allergies (e.g., to penicillin, cephalosporins), insect sting allergies; psoriasis and inflammatory dermatoses such as dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); spondyloarthropathies; scleroderma; respiratory allergic diseases such as asthma, allergic rhinitis;
      • autoimmune diseases, such as arthritis (e.g., rheumatoid arthritis, psoriatic arthritis), multiple sclerosis, systemic lupus erythematosus, myasthenia gravis, juvenile onset diabetes, glomerulonephritis and other nephritides, autoimmune thyroiditis, Behcet's disease;
      • graft rejection (e.g., in transplantation), including allograft rejection or graft-versus-host disease;
      • other diseases or conditions in which undesirable inflammatory responses are to be inhibited can be treated, including, but not limited to, atherosclerosis, restenosis, myositis (including polymyositis, dermatomyositis).
    Modes of Administration
  • The compound can be administered as a neutral compound or as a salt. Salts of compounds containing an amine or other basic group can be obtained, for example, by contacting the compound with a suitable organic or inorganic acid, such as hydrogen chloride, hydrogen bromide, acetic acid, perchloric acid and the like. Compounds with a quaternary ammonium group also contain a counteranion such as chloride, bromide, iodide, acetate, perchlorate and the like. Other examples of such salts include hydrochlorides, hydrobromides, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, tartrates [e.g. (+)-tartrates, (−)-tartrates or mixtures thereof including racemic mixtures], succinates, benzoates and salts with amino acids such as glutamic acid.
  • Salts of compounds containing a carboxylic acid or other acidic functional group can be prepared by reacting with a suitable base. Such a pharmaceutically acceptable salt may be made with a base which affords a pharmaceutically acceptable cation, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and magnesium), aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N,N′-dibenzylethylenediamine, 2-hydroxyethylamine, bis-(2-hydroxyethyl)amine, tri-(2-hydroxyethyl)amine, procaine, dibenzylpiperidine, N-benzyl-β-phenethylamine, dehydroabietylamine, N,N′-bisdehydroabietylamine, glucamine, N-methylglucamine, collidine, quinine, quinoline, and basic amino acid such as lysine and arginine. These salts may be prepared by methods known to those skilled in the art.
  • Salts of acidic functional groups contain a counteraction such as sodium, potassium and the like.
  • Certain compounds described herein and their salts may also exist in the form of solvates, for example hydrates, and the present invention includes each solvate and mixtures thereof.
  • Certain compounds described herein may exist in different stable conformational forms which may be separable. Torsional asymmetry due to restricted rotation about an asymmetric single bond, for example because of steric hindrance or ring strain, may permit separation of different conformers. The present invention includes each conformational isomer of these compounds and mixtures thereof.
  • Certain compounds described herein may exist in zwitterionic form. The present invention includes each zwitterionic form of these compounds and mixtures thereof.
  • Certain compounds described herein and their salts may exist in more than one crystal form. Polymorphs of these compounds form part of this invention and may be prepared by crystallization of the compound under different conditions, for example, by using different solvents or different solvent mixtures for recrystallization; by crystallization at different temperatures; or by various modes of cooling, ranging from very fast to very slow cooling during crystallization. Polymorphs may also be obtained by heating or melting the compound followed by gradual or fast cooling. The presence of polymorphs may be determined by solid probe nmr spectroscopy, it spectroscopy, differential scanning calorimetry, powder X-ray diffraction or such other techniques.
  • The language an “effective amount” or “pharmaceutically effective amount” is intended to include an amount which is sufficient to ameliorate a disease or condition and prevent its further progression or ameliorate the symptoms associated with the disease or condition. Such an amount can be administered prophylactically to a patient thought to be susceptible to development of a disease or condition. Such amount when administered prophylactically to a patient can also be effective to prevent or lessen the severity of the mediated condition. Such an amount is intended to include an amount which is sufficient to modulate a CCR9 receptor-mediated disease or condition. Conditions mediated by CCR9 receptors include all of the diseases or conditions described herein. Although the amount to be administered to a subject will, of course, be determined by a physician, in the light of all the relevant circumstances, an “effective amount” typically ranges between about 0.01 mg/kg/day to about 100 mg/kg/day, preferably between about 0.5 mg/kg/day to about 50 mg/kg/day.
  • The compounds described herein, and the pharmaceutically acceptable salts, solvates and hydrates thereof, have valuable pharmacological properties and can be used in pharmaceutical preparations containing the compound or pharmaceutically acceptable salts thereof, in combination with a pharmaceutically acceptable carrier or diluent. They are useful as therapeutic substances in preventing or treating diseases mediated by CCR9 receptors, such as inflammatory diseases, in human or non-human animals. Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions. The active compound will be present in such pharmaceutical compositions in amounts sufficient to provide the desired dosage amount in the range described herein. Techniques for formulation and administration of the compounds of the instant invention can be found in Remington: the Science and Practice of Pharmacy, 19th edition, Mack Publishing Co., Easton, Pa. (1995).
  • For oral administration, the compound or salts thereof can be combined with a suitable solid or liquid carrier or diluent to form capsules, tablets, pills, powders, syrups, solutions, suspensions and the like.
  • The tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacias, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid, a lubricant such as magnesium stearate; and a sweetening agent such as sucrose lactose or saccharin. When a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor.
  • The active compounds can also be administered intranasally as, for example, liquid drops or spray. For oral or nasal inhalation, the compounds for use according to the present invention are conveniently delivered in the form of a dry powder inhaler, or an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • For parental administration the compounds of the present invention, or salts thereof can be combined with sterile aqueous or organic media to form injectable solutions or suspensions. For example, solutions in sesame or peanut oil, aqueous propylene glycol and the like can be used, as well as aqueous solutions of water-soluble pharmaceutically-acceptable salts of the compounds. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that each syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against any contamination. The carrier can be solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g. glycerol, propylene glycol and liquid polyethylene glycol), propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils. The injectable solutions prepared in this manner can then be administered intravenously, intraperitoneally, subcutaneously, or intramuscularly, with intramuscular administration being preferred in humans.
  • The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • In addition, to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation, for example, subcutaneously or intramuscularly or by intramuscular injection. Thus, for example, as an emulsion in an acceptable oil, or ion exchange resins, or as sparingly soluble derivatives, for example, as sparingly soluble salts.
  • The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated.
  • The compositions are formulated and administered in the same general manner as detailed herein. The compounds of the instant invention may be used effectively alone or in combination with one or more additional active agents depending on the desired target therapy. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound described herein and one or more additional active agents, as well as administration of the compound and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound described herein or salt thereof can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations. Where separate dosage formulations are used, a compound of the invention and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
  • An effective amount of the compounds described herein can be used for the preparation of a medicament useful for treating a disease mediated by CCR9 receptors, such as an inflammatory disease, and for treating, preventing or reducing the risk of developing a disease mediated by CCR9 receptors, such as an inflammatory disease, in mammals, particularly in humans.
  • Preferably compounds of the invention or pharmaceutical formulations containing these compounds are in unit dosage form for administration to a mammal. The unit dosage form can be any unit dosage form known in the art including, for example, a capsule, an IV bag, a tablet, or a vial. The quantity of active ingredient (viz., a compound of Structural Formula I or salts thereof) in a unit dose of composition is an effective amount and may be varied according to the particular treatment involved. It may be appreciated that it may be necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of administration which may be by a variety of routes including oral, aerosol, rectal, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal and intranasal.
  • Pharmaceutical formulations of the invention are prepared by combining (e.g., mixing) an effective amount of a compound of the invention together with a pharmaceutically acceptable carrier or diluent. The present pharmaceutical formulations are prepared by known procedures using well known and readily available ingredients.
  • In making the compositions of the present invention, the active ingredient will usually be admixed with a carrier, or diluted by a carrier, or enclosed within a carrier which may be in the form of a capsule, sachet, paper or other container. When the carrier serves as a diluent, it may be a solid, lyophilized solid or paste, semi-solid, or liquid material which acts as a vehicle, or can be in the form of tablets, pills, powders, lozenges, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), or ointment, containing, for example, up to 10% by weight of the active compound. The compounds of the present invention are preferably formulated prior to administration.
  • For the pharmaceutical formulations any suitable carrier known in the art can be used. In such a formulation, the carrier may be a solid, liquid, or mixture of a solid and a liquid.
  • Advantageously, compositions containing the compounds of the invention or the salts thereof may be provided in dosage unit form, each dosage unit containing from about 5% to about 95%, preferably about 20% to about 80%, of a compound of the invention, although it will, of course, readily be understood that the amount of the compound or compounds actually to be administered will be determined by a physician, in the light of all the relevant circumstances.
  • Powders and tablets preferably contain from about 1 to about 99 weight percent of the active ingredient which is the novel compound of this invention. Suitable solid carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, low melting waxes, and cocoa butter.
  • The following pharmaceutical formulations 1 through 8 are illustrative only and are not intended to limit the scope of the invention in any way. “Active Ingredient”, refers to a compound according to Structural Formula I or salts thereof.
  • Formulation 1
  • Hard gelatin capsules are prepared using the following ingredients:
  • Quantity
    (mg/capsule)
    Active Ingredient 250
    Starch, dried 200
    Magnesium stearate 10
    Total 460 mg
  • Formulation 2
  • A tablet is prepared using the ingredients below:
  • Quantity
    (mg/tablet)
    Active Ingredient 250
    Cellulose, microcrystalline 400
    Silicon dioxide, fumed 10
    Stearic acid 5
    Total 665 mg
  • The components are blended and compressed to form tablets each weighing 665 mg
  • Formulation 3
  • An aerosol solution is prepared containing the following components:
  • Weight
    Active Ingredient 0.25
    Ethanol 25.75
    Propellant 22 (Chlorodifluoromethane) 74.00
    Total 100.00
  • The Active Ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 30° C. and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remainder of the propellant. The valve units are then fitted to the container.
  • Formulation 4
  • Tablets, each containing 60 mg of Active ingredient, are made as follows:
  • Active Ingredient 60 mg
    Starch 45 mg
    Microcrystalline cellulose 35 mg
    Polyvinylpyrrolidone (as 10% solution in water)  4 mg
    Sodium carboxymethyl starch 4.5 mg 
    Magnesium stearate 0.5 mg 
    Talc  1 mg
    Total 150 mg 
  • The Active Ingredient, starch and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly. The aqueous solution containing polyvinylpyrrolidone is mixed with the resultant powder, and the mixture then is passed through a No. 14 mesh U.S. sieve. The granules so produced are dried at 50° C. and passed through a No. 18 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate and talc, previously passed through a No. 60 mesh U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
  • Formulation 5
  • Capsules, each containing 80 mg of Active Ingredient, are made as follows:
  • Active Ingredient 80 mg
    Starch 59 mg
    Microcrystalline cellulose 59 mg
    Magnesium stearate  2 mg
    Total 200 mg 
  • The Active Ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 45 mesh U.S. sieve, and filled into hard gelatin capsules in 200 mg quantities.
  • Formulation 6
  • Suppositories, each containing 225 mg of Active Ingredient, are made as follows:
  • Active Ingredient   225 mg
    Saturated fatty acid glycerides 2,000 mg
    Total 2,225 mg
  • The Active Ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool.
  • Formulation 7
  • Suspensions, each containing 50 mg of Active Ingredient per 5 mL dose, are made as follows:
  • Active Ingredient 50 mg
    Sodium carboxymethyl cellulose 50 mg
    Syrup 1.25 mL
    Benzoic acid solution 0.10 mL
    Flavor q.v.
    Color q.v.
    Purified water to total 5 mL
  • The Active Ingredient is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form a smooth paste. The benzoic acid solution, flavor and color are diluted with a portion of the water and added, with stirring. Sufficient water is then added to produce the required volume.
  • Formulation 8
  • An intravenous formulation may be prepared as follows:
  • Active Ingredient 100 mg
    Isotonic saline 1,000 mL
  • The solution of the above materials generally is administered intravenously to a subject at a rate of 1 mL per minute.
  • EXAMPLES
  • Figure US20100324093A1-20101223-C00032
  • The sulfonamides of the invention can be synthesized by treating a protected aromatic amine (XXVI) or a protected heteroaromatic amine (XXVI), such as an aminobenzene, an aminopyridine, an aminopyrimidine, an aminopyridazine, an aminopyrazine, an aminotriazine or an aminotetrazine, with an alkyl lithium. Typically, the amino group of the aromatic amine or heteroaromatic amine is protected with protecting group which is stable under basic condition, such as a tert-butoxycarbonyl (BOC) group. The aromatic amine or heteroaromatic amine is dissolved in an aprotic solvent, preferably an ether. The solution is then cooled to about −50° C. to about −100° C. and about 1 equ. to about 2.5 equ. of alkyl lithium (e.g., n-butyl lithium, s-butyl lithium or t-butyl lithium) is added slowly to the reaction mixture. The temperature of the reaction mixture is allowed to increase to about −25° C. to about 25° C. After about 5 min. to about 45 min., about 1 equ. to about 1.5 equ. of a Weinreb amide (XXVII), such as a N-methoxy-N-methyl-benzamide or a N-methoxy-N-methyl-isonicotinamide, dissolved in an aprotic solvent is added to the reaction mixture. The reaction is allowed to continue stirring until the protected aromatic amine (XXVI) or protected heteroaromatic amine (XXVI) is consumed (typically, for about 0.5 hr to about 3 hr at about 15° C. to about 35° C.). The reaction is monitored by thin layer chromatography (tic) to determine when the aromatic amine (XXVI) or heteroaromatic amine (XXVI) has been consumed. When the reaction is complete, it is quenched with an acidic aqueous solution to yield compound XXVIII.
  • After the amino group of compound XXVIII has been deprotected to yield compound XXIX, compound XXIX is converted to a sulfonamide by using one of two alternative methods. In method 1, compound XXIX is dissolved in an aprotic solvent and the solution is cooled to about −25° C. to about 10° C. and about 1 equ. to about 1.5 equ. of NaH is added to the mixture. After about 1 min. to about 15 min., about 1 equ. to about 1.5 equ. of sulfonyl chloride (XXX) dissolved in an aprotic solvent is added to the reaction mixture. An additional, amount of up to about 0.5 equ. NaH may be added to the reaction mixture. The reaction is stirred at about 15° C. to about 30° C. until the reaction is complete as indicated by tlc (typically, about 1 hr to about 3 hr). The reaction is quench by addition of water to yield sulfonamide (XXXI).
  • In the second method of forming sulfonamide (XXXI), compound XXIX is dissolved in an aprotic solvent and the mixture is cooled to about −10° C. to about 10° C. An excess amount of sodium bis(trimethylsilyl)amide (typically, about 1.5 equ. to about 3 equ.) in an aprotic solvent is added to the solution, and the reaction is allowed to stir for about 10 min. to about 45 min. A solution of about 1 equ. to about 2 equ. of sulfonyl chloride (XXX) in an aprotic solvent is added to the reaction mixture and the reaction is allowed to warm up to about 15° C. to about 30° C. and is stirred for about 6 hr to about 24 hr. The reaction is quenched with an acidic water solution to yield sulfamide (XXXI).
  • II. General Synthesis of N-(2-Benzoyl-Phenyl)-Arylsulfonamides and N-(2-Benzoyl-Phenyl)-Heteroarylsulfonamides
  • Figure US20100324093A1-20101223-C00033
  • Procedure A Synthesis of Weinreb Amides
  • A substituted anthranilic acid (24 mmol) was dissolved in acetonitrile (200 mL). 1.05 equivalents of N,O dimethylhydroxylamine hydrochloride, 1.05 equivalents of EDC, 0.05 equivalents of dimethylaminopyridine, and 1.0 equivalent of triethylamine were added and the reaction was stirred at room temperature overnight. The acetonitrile was removed by rotary evaporation and the residue was partitioned between ethyl acetate and water. The organic layer was washed with brine then concentrated to a residue. The residue was chromatographed on silica gel (ethyl acetate as eluent) to give the product. Typical yields are 70-90%.
  • The following compounds were prepared using Procedure A:
  • Example 1 2-Amino-5-bromo-N-methoxy-N-methyl-benzamide
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 259 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 7.5 (s, 1H) δ 7.23 (d, 1H) δ 6.58 (d, 1H) δ 4.68 (br s, 2H) δ 3.55 (s, 3H) δ 3.41 (s, 3H).
  • Example 2 2-Amino-N-methoxy-N-methyl-5-nitro-benzamide
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 226 (M+H+). 1H NMR (CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    8.5 (s, 1H),
    Figure US20100324093A1-20101223-P00002
    8.1 (d, 1H)
    Figure US20100324093A1-20101223-P00001
    6.7 (d, 1H)
    Figure US20100324093A1-20101223-P00001
    3.6 (s, 3H)
    Figure US20100324093A1-20101223-P00001
    3.4 (s, 3H).
  • Example 3 2-Amino-5-chloro-N-methoxy-N-methyl-benzamide
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 215. (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    7.4 (s, 1H)
    Figure US20100324093A1-20101223-P00001
    7.15 (m, 1H)
    Figure US20100324093A1-20101223-P00001
    6.65 (d, 1H)
    Figure US20100324093A1-20101223-P00001
    3.6 (s, 3H)
    Figure US20100324093A1-20101223-P00001
    3.28 (s, 3H).
  • Procedure B General synthesis of substituted 2-aminobenzophenones
  • A substituted Weinreb amide (10 mmol) and a substituted bromobenzene (10 mmol) were dissolved in THF under nitrogen. The reaction mixture was cooled to −100° C. using a liquid nitrogen/diethyl ether bath. n-BuLi (21 mmol, 1.6 M in cyclohexanes) was introduced via syringe over a period of twenty minutes. The reaction mixture was allowed to warm to −70° C. and then quenched by adding of 21 mL of 1 N HCl. The reaction mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organic layer was washed with saturated sodium chloride, then concentrated to a residue. The residue was chromatographed on silica gel using 1:1 ethyl acetate:hexanes to give the product. Typical yields are 30-60%.
  • The following compounds were prepared using Procedure B:
  • Example 4 (2-Amino-5-bromo-phenyl)-phenyl-methanone
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 276 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 7.2-7.8 (m, 6H) δ 6.85 (d, 2H) δ 6.2 (br s, 2H).
  • Example 5 (2-Amino-5-nitro-phenyl)-(3-trifluoromethyl-phenyl)-methanone
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 311 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    7.6-8.4 (m, 5H)
    Figure US20100324093A1-20101223-P00001
    6.75 (d, 2H)
    Figure US20100324093A1-20101223-P00001
    7.0 (br s, 2H).
  • Example 6 (2-Amino-5-chloro-phenyl)-m-tolyl-methanone
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 246. (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    7.25-7.5 (m, 5H)
    Figure US20100324093A1-20101223-P00001
    6.8 (d, 2H)
    Figure US20100324093A1-20101223-P00001
    2.4 (s, 3H).
  • Procedure C General synthesis of N-(2-benzoyl-phenyl)-benzenesulfonamides
  • A substituted 2-aminobenzophenone (1 mmol) was dissolved in 5 mL of pyridine. A substituted sulfonyl chloride (1.3 mmol) and dimethlaminopyridine (10 mg) were added to the 2-aminobenzophenone solution. The reaction mixture was heated to 110° C. for 2 hours. The solvent was evaporated under a stream of nitrogen and the product was isolated by reverse phase HPLC using the method given below. Typical yields are 60-80%.
  • General Method for HPLC Purification of N-(2-benzoyl-phenyl)-benzenesulfonamides:
  • A crude N-(2-benzoyl-phenyl)-benzenesulfonamides was dissolved in 1 mL of MeOH (or MeOH/DCM) and filtered through a 0.45 micron filter. The solution was then injected (50-75 mg/injection) to a HPLC/MS system equipped with Waters 2700 Sample Manager auto-injector, Waters 600 Controller and Pumps, Waters 996 Diode Array detector, Micro Mass Platform LCZ mass spectrometer and Gilson FC-204 fraction collector. Solvents A and B were used for gradient elution of the purified compound using Phenomenex Luna 15 micron, C18(2) 100A, 100×21.2 mm column at 20 mL/min flow rate.
  • Solvent A: 99% Water/1% CH3CN/0.1% Formic Acid Solvent B: 95% CH3CN/5% Water/0.1% Formic Acid
  • The gradients were programmed according to their analytical scale HPLC retention time (tana) which was obtained by running a linear gradient of 0% B to 100% B in A in 3.5 min on a Phenomenex Luna 5 micron C18(2) 50×4.6 mm column at 3.5 mL/min.
  • Linear mixing of 0% of B to X % of B in A was used according to the following formula with some adjustments for some compounds.

  • X=t ana×95/3.5+15
  • About 15 mL to 25 mL size fractions were collected based on mass triggered collection. They were combined according to their purities based on mass spectral pattern of the fractions.
  • The following examples were prepared using Procedure C:
  • Example 7 N-[4-Chloro-2-(2-chloro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00034
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 451 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.0 (s, 1H) δ 8.0-8.4 (dd, 4H) δ 7.8 (d, 1H) δ 7.38-7.58 (m, 6H).
  • Example 8 Thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide
  • Figure US20100324093A1-20101223-C00035
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 378 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.0 (s, 1H) δ 7.85 (d, 1H) δ 7.4-7.7 (m, 9H) δ 6.85 (d, 1H).
  • Example 9 N-(2-Benzoyl-4-chloro-phenyl)-4-iodo-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00036
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 498 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.7 (s, 1H) δ 7.3-7.8 (m, 12H).
  • Example 10 N-(2-Benzoyl-4-chloro-phenyl)-4-chloro-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00037
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 406 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.65 (s, 1H) δ 7.2-7.8 (m, 12H).
  • Example 11 N-(2-Benzoyl-4-chloro-phenyl)-4-tert-butyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00038
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 428 (M+H+). 1H NMR (CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.9 (s, 1H) δ 7.2-7.8 (m, 12H), δ 1.18 (s, 9H).
  • Example 12 N-(2-Benzoyl-4-chloro-phenyl)-4-propyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00039
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 414 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.9 (s, 1H) δ 7.0-7.8 (m, 12H), δ 2.42 (m, 2H) δ 1.5 (m, 2H) δ 0.92 (m, 3H).
  • Example 13 N-(2-Benzoyl-4-chloro-phenyl)-4-ethyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00040
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 400 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.9 (s, 1H) δ 7.0-7.8 (m, 12H), δ 2.42 (t, 2H) δ 0.92 (d, 3H).
  • Example 14 N-(2-Benzoyl-4-nitro-phenyl)-4-ethyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00041
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 411 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.6 (s, 1H) δ 8.2-8.45 (m, 2H) δ 7.2-7.9 (m, 10H), δ 2.6 (q, 2H) δ 1.05 (t, 3H).
  • Example 15 N-(2-Benzoyl-4-chloro-phenyl)-4-isopropyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00042
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 414 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.85 (s, 1H) δ 7.0-7.8 (m, 12H), δ 2.8 (m, 1H) δ 1.03 (d, 6H).
  • Example 16 N-(2-Benzoyl-4-bromo-phenyl)-4-ethyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00043
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 444 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    9.8 (s, 1H),
    Figure US20100324093A1-20101223-P00001
    7.38-7.8 (m, 10H)
    Figure US20100324093A1-20101223-P00001
    7.08 (d, 2H)
    Figure US20100324093A1-20101223-P00001
    2.47 (q, 2H)
    Figure US20100324093A1-20101223-P00001
    1,1 (t, 3H).
  • Example 17 N-(2-Benzoyl-4-bromo-phenyl)-4-methoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00044
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 446 (M+H+). 1H NMR Bruker 300 MHz, (CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    9.61 (s, 1H)
    Figure US20100324093A1-20101223-P00001
    7.2-7.8 (m, 10H),
    Figure US20100324093A1-20101223-P00001
    6.64 (d, 2H)
    Figure US20100324093A1-20101223-P00001
    3.78 (s, 3H).
  • Example 18 N-(2-Benzoyl-4-bromo-phenyl)-4-isopropyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00045
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 459 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    9.9 (s, 1H),
    Figure US20100324093A1-20101223-P00001
    7.28-7.8 (m, 10H)
    Figure US20100324093A1-20101223-P00001
    7.16 (d, 2H),
    Figure US20100324093A1-20101223-P00001
    2.8 (m, 1H)
    Figure US20100324093A1-20101223-P00001
    1.08 (d, 1H),
  • Example 19 4-Ethyl-N-[4-nitro-2-(3-trifluoromethyl-benzoyl)-phenyl]-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00046
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 479 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    10.9 (s, 1H),
    Figure US20100324093A1-20101223-P00001
    8.4 (m, 2H)
    Figure US20100324093A1-20101223-P00001
    7.6-8.0 (m, 6H)
    Figure US20100324093A1-20101223-P00001
    7.28 (m, 2H)
    Figure US20100324093A1-20101223-P00001
    2.62 (q, 2H)
    Figure US20100324093A1-20101223-P00001
    1.09 (t, 3H).
  • Example 20 N-[4-Chloro-2-(3-methyl-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00047
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 414 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    9.8 (s, 1H)
    Figure US20100324093A1-20101223-P00001
    7.0-7.9 (m, 11H)
    Figure US20100324093A1-20101223-P00001
    2.48 (q, 2H)
    Figure US20100324093A1-20101223-P00001
    2.39 (s, 3H)
    Figure US20100324093A1-20101223-P00001
    1.13 (t, 3H).
  • Example 21 N-(2-Benzoyl-4-bromo-phenyl)-4-chloro-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00048
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 450 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm)
    Figure US20100324093A1-20101223-P00001
    9.7 (s, 1H),
    Figure US20100324093A1-20101223-P00001
    7.2-8.0 (m, 12H).
  • Procedure D Alternate synthesis of N-(2-benzoyl-phenyl)-benzenesulfonamides
  • A solution of substituted 2-aminobenzophenone (0.5 mmol), sulfonyl chloride (0.7 mmol), and N-methylmorpholine (0.9 mmol) in dichloromethane (5 mL) was stirred at 40° C. for 20 hours. The mixture was cooled to room temperature, washed with water (2×5 mL), brine (5 mL) dried over Na2SO4, filtered, and purified by silica gel chromatography using dichloromethane:hexanes (1:1) to afford the desired N-(2-benzoyl-phenyl)-benzenesulfonamides.
  • The following examples were prepared using Procedure D:
  • Example 22 N-(2-Benzoyl-4-chloro-phenyl)-4-ethoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00049
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 416 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 1.30-1.40 (t, 3H), δ 3.85-3.95 (m, 2H), δ 6.68-6.70 (d, 2H), δ 7.30-7.65 (m, 9H), δ 7.75-7.77 (d, H), δ 9.65 (s, H).
  • Example 23 N-(2-Benzoyl-4-chloro-phenyl)-4-propoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00050
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 430 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 0.99-1.10 (t, 3H), δ 1.70-1.80 (m, 2H), δ 3.70-3.75 (t, 2H), δ 6.68-6.70 (d, 2H), δ 7.30-7.65 (m, 9H), δ 7.75-7.77 (d, H), δ 9.65 (s, H).
  • Example 24 N-(2-Benzoyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00051
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 430 (M+H+). 1H-NMR (CDCl3, Bruker 300 MHz): δ 1.25-1.26 (d, 6H), δ 4.30-4.48 (m, H), δ 6.68-6.70 (d, 2H), δ 7.30-7.65 (m, 9H), δ 7.75-7.77 (d, H), δ 9.70 (s, H).
  • Example 25 N-(2-Benzoyl-4-chloro-phenyl)-4-butoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00052
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 444 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 0.95-1.00 (t, 3H), δ 1.40-1.60 (m, 2H), δ 1.70-1.80 (m, 2H), δ 3.75-3.80 (t, 2H), δ 6.68-6.70 (d, 2H), δ 7.30-7.65 (m, 9H), δ 7.75-7.77 (d, H), δ 9.70 (s, H).
  • Example 26 N-(2-Benzoyl-4-chloro-phenyl)-4-benzyloxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00053
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 478 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 4.93 (s, 2H), δ6.75-6.80 (d, 2H), δ 7.335-7.55 (m, 11H), δ 7.60-7.70 (m, 3H), δ 7.78-7.80 (d, H), δ 9.70 (s, H).
  • Example 27 N-(2-Benzoyl-4-chloro-phenyl)-4-phenoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00054
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 464 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ (ppm)=6.78-6.80 (d, 2H), 6.90-6.93 (d, 2H), 7.20-7.24 (t, H), 7.33-7.45 (m, 3H), 7.45-7.53 (m, 5H), 7.60-7.70 (m, 3H), 7.75-7.77 (d, H), 9.78 (s, H).
  • III. General Synthesis of 2-Amino-Pyridophenone-Sulfonamides
  • Figure US20100324093A1-20101223-C00055
  • Procedure E BOC Protection of Substituted 2-Aminopyridines)
  • To a mixture of 2-amino-5-substituted-pyridine (0.10 mol), 4-dimethylaminopyridine (DMAP, 1.22 g, 0.01 mol), and triethylamine (TEA, 20 mL, 0.15 mol) in dichloromethane (80 mL) and DMF (4 mL) at 0° C. was added a solution of di-t-butyl dicarbonate (28.4 g, 0.13 mol) in dichloromethane (20 mL). The mixture was stirred at room temperature overnight. The mixture was washed with water (3×100 mL), brine (100 mL), dried with anhydrous Na2SO4, and filtered. The filtrate was concentrated in vacuo to afford a solid which was collected by filtration and washed with small amount of DCM to give the expected product in 40-60% yield.
  • The following example was prepared using Procedure E:
  • Example 28 (5-Chloro-pyridin-2-yl)-carbamic acid tert-butyl ester
  • hu 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) 1.54 (s, 9H), 7.59-7.64 (dd, H), 7.94-7.97 (d, H), 8.24-8.25 (dd, H), 8.39 (b, H). LC-MS showed the product to be >95% pure. The molecular ion was not observed but the fragment corresponding to loss of the t-butyl group was evident (M.W. of 173/175).
  • Procedure F Synthesis 2-aminopyridin-3-yl)-phenyl-methanones or 2-aminopyridin-3-yl)-pyridin-4-yl-methanones
  • A solution of BOC protected 5-substituted 2-aminopyridine (10 mmol) in anhydrous tetrahydrofuran (THF, 5 mL) was cooled to −78° C. under nitrogen atmosphere with stirring. A solution of n-butyl lithium in hexane (2.5 M, 8.8 mL) was added dropwise, and the mixture was stirred at 0° C. for 10 min. A Weinreb amide (12 mmol) was prepared as in Procedure A and was dissolved in anhydrous THF (5 mL) and added to the reaction mixture. The reaction mixture was warmed to room temperature and stirred for 2 hours. The reaction was added to an ice-0.18 M aqueous citric acid solution (100 g-130 mL) mixture, then extracted with ethyl acetate (EtOAc, 150 mL and 50 mL). The combined organic layer was washed with water (2×100 mL), brine (100 mL), dried over anhydrous Na2SO4, and filtered. The crude material concentrated from the filtrate was purified by flash chromatography to give a solid product. This compound was heated in 4 N HCl/dioxane (6 mL) at 100° C. for 2 hours, then cooled to room temperature. The mixture was evaporated in vacuo and the residue was taken up into a saturated aqueous NaHCO3 solution (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layer was washed with water (2×50 mL), then with brine (50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was evaporated and dried in vacuum to afford a pale yellow solid product in yields of 40-60% for the two steps.
  • The following examples were prepared using Procedure F:
  • Example 29 (2-Amino-5-chloro-pyridin-3-yl)-pyridin-4-yl-methanone
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 234 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 6.96 (b, 2H), δ 7.40-7.44 (m, 2H), δ 7.62-7.63 (d, H), δ 8.25-8.26 (d, H), δ 8.81-8.84 (m, 2H).
  • Example 30 (2-Amino-5-chloro-pyridin-3-yl)-phenyl-methanone
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 233 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 6.69-6.84 (br s, 2H), 7.48-7.54 (m, 2H), 7.57-7.65 (m, 3H), 7.74-7.75 (d, H), 8.22-8.23 (d, H).
  • Procedure G Alternate Synthesis of Sulfonamides Using NaH
  • To a solution of a 2-aminopyridophenone (0.5 mmol) in dry DMF (1 mL) at 0° C. was added NaH (60%, 20 mg, 0.5 mmol). The mixture was stirred for 5 min. The a sulfonyl chloride (0.55 mmol) in dry DMF (1 mL) was added, and the mixture stirred at 0° C. for 10 min. The second portion of NaH (0.25 mmol) was added, and the mixture stirred at room temperature for 2 hours. The mixture was quenched with ice water (20 mL) and extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (2×10 mL), brine (10 mL), dried over anhydrous Na2SO4, filtered, and evaporated in vacuo to give crude product. The crude product was purified by chromatography using DCM:EtOAc (100:0 to 98:2) or hexane:EtOAc (9:1) as eluent to afford the purified product in 10-24% yield.
  • The following examples were prepared using Procedure G:
  • Example 31 N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00056
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 431 (M+H+) 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 1.34-1.36 (d, 6H), δ 4.57-4.70 (m, H), δ 6.91-6.98 (d, 2H), δ 7.50-7.70 (m, 5H), δ 7.82-7.83 (d, H), δ 8.08-8.12 (d, 2H), δ 8.38-8.39 (d, H), δ 10.70 (s, H). Elemental analysis: C=58.26%/cal. 58.53%, H=4.65%/cal. 4.44%, N=6.26%/cal. 6.50%.
  • Example 32 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00057
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 417 (M+H+); 1H NMR (CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.90 (1H, s), δ 8.80 (2H, s), δ 7.79 (1H, d), δ 6.76 (2H, d), δ 7.51 (1H, d), δ 7.33-7.20 (3H, m), δ 6.75 (2H, d), δ 3.90 (2H, m), δ 1.5 (3H, m).
  • Example 33 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00058
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 415 (M+H+); 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.06 (1H, s), δ 8.77 (1H, s), δ 7.79 (1H, d), δ 7.70-7.62 (3H, m), δ 7.56-7.51 (1H, m), δ 7.33-7.16 (5H, m), δ 1.19 (6H, m), δ 2.88 (1H, m).
  • Example 34 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isobutyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00059
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 429 (M+H+); 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.06 (1H, s), δ 7.77 (1H, s), δ 7.67-7.61 (3H, m), δ 7.58-7.47 (1H, m), δ 7.33-7.24 (3H, m), δ 7.17-7.08 (3H, m), δ 2.42 (2H, m), δ 0.86 (6H, m).
  • Another example of a compound prepared using Procedure G is N-[4-chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethyl-benzenesulfonamide. LC-MS showed the product to be >95% pure and to have the expected M.W. of 401 (M+H+).
  • Procedure H Alternate Synthesis of Sulfonamides Using Na(TMS)2N)
  • To a suspension of a 2-aminopyridophenone (1 mmol) in dry DMF (5 mL) at 0° C. was added a solution of sodium bis(trimethylsilyl)amide in THF (1M, 2.2 mL). This mixture was stirred at 0° C. for 20 min. A solution of a sulfonyl chloride (1.3 mmol) in dry DMF (2 mL) was added, and the reaction mixture was stirred at room temperature overnight. The reaction was quenched with ice water (60 mL), neutralized with 0.18 M aqueous citric acid solution (3 mL) to about pH 7.5, then extracted with EtOAc (60 mL, 2×20 mL). The combined organic layers were washed with water (2×50 mL), brine (50 mL), dried over anhydrous Na2SO4, filtered, and evaporated in vacuo to give a crude product. The crude product was purified by chromatography using DCM:EtOAc (6:4) as elute to afford a solid product in 20-25% yield.
  • The following examples were prepared using Procedure H:
  • Example 35 N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-ethoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00060
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 418 (M+H+). 1H-NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 1.40-1.44 (t, 3H), δ4.03-4.12 (m, 2H), δ 6.93-6.96 (d, 2H), δ 7.39-7.42 (d, 2H), δ 7.70-7.72 (d, H), δ 8.08-8.11 (d, 2H), δ 8.39-8.40 (d, H), δ 8.84-8.86 (d, 2H), δ 10.73 (s, H).
  • Example 36 N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00061
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 432 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm): δ 1.35-1.36 (d, 6H), δ 4.57-4.67 (m, H), δ 6.92-6.95 (d, 2H), δ 7.42-7.44 (d, 2H), δ 7.72-7.73 (d, H), δ 8.08-8.11 (d, 2H), δ 8.41-8.42 (d, H), δ 8.84-8.87 (d, 2H), δ 10.74 (s, H
  • IV. Synthesis of (2-Amino-5-Chloro-Phenyl)-Pyridin-3-yl-Methanones
  • Figure US20100324093A1-20101223-C00062
  • Procedure I Alternate synthesis of (2-amino-5-choro-phenyl)-pyridin-3-yl-methanones using BCl3
  • To a solution of 85 mL BCl3 in 80 mL 1,1,2,2-tetrachloroethane, stirred in an ice bath, was added a solution of 4 g of p-chloroaniline in 50 mL of tetrachloroethane. After 5 min, 3 g of 3-cyanopyridine is added followed by 7 g of aluminum chloride. The mixture was stirred at room temperature for 20 min and then heated at reflux for 6 hr. After cooling, 25 mL of 3N HCl was added and refluxing continued for 1 hr. After cooling, the filtrate was concentrated, made basic with 6N NaOH, then extraction with dichloromethane. The organic solution was dried over MgSO4 and concentrated. The residue was flash chromatographed on silica gel (Hexane/EtOAc=2:1) to give a 10-20% yield of product.
  • The following Example was prepared using Procedure I:
  • Example 37 (2-Amino-5-chloro-phenyl)-pyridin-3-yl-methanone
  • Figure US20100324093A1-20101223-C00063
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 233. (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 8.95 (s, 1H), δ 8.80 (d, 1H), δ 8.18 (d, 1H), δ 7.40-7.45 (m, 1H), δ 7.20-7.30 (m, 2H) δ 7.10 (s, 1H), δ 6.30 (br s, 2H).
  • (2-Amino-5-choro-phenyl)-pyridin-3-yl-methanones prepared using the method depicted in Scheme III can be converted to the corresponding aryl sulfonamide or heteroaryl sulfonamide using Procedure G or H. The following example was prepared using the method depicted in Scheme III followed by Procedure G or H:
  • Example 38 N-[4-Chloro-2-(pyridine-3-carbonyl)-phenyl]-4-methyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00064
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 387 (M+H+); 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 9.66 (s, 1H), δ 7.87-7.76 (m, 3H), δ 7.56-7.50 (m, 4H), δ 7.43-7.40 (m, 1H), δ 7.28 (s, 1H), δ 7.08 (d, 2H), δ 3.50 (s, 3H).
  • V. Synthesis of (2-amino-5-chloro-phenyl)-pyridin-4-yl-methanones
  • Figure US20100324093A1-20101223-C00065
  • Procedure J Pivaloyl Protection of 4-Chloro Aniline
  • To a mixture of 2.3 g of 4-chloroaniline and 3 mL of triethylamine in 20 mL of dichloromethane was added 2.3 g of trimethylacetyl chloride. The reaction mixture was stirred overnight at room temperature, then washed with 1 N HCl. The organic layer was separated and dried over MgSO4. Removal of the solvent afforded 4.5 g of pure product.
  • The following examples were prepared using Procedure J:
  • Example 39 N-(5-Chloro-pyridin-2-yl)-2,2-dimethyl-propionamide
  • Figure US20100324093A1-20101223-C00066
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 213 (M+H+).
  • Example 40 N-(4-Chloro-phenyl)-2,2-dimethyl-propionamide
  • Figure US20100324093A1-20101223-C00067
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 212. (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ10.51 (s, 1H), δ 7.25 (d, 2H), δ7.15 (d, 2H), δ1.51 (s, 9H).
  • Procedure K
  • A solution of 2 g of pivaloyl protected 4-chloroaniline in 50 mL of THF was cooled to −78° C. under nitrogen. A solution of n-butyl lithium (14 mL of 2 N in hexane) was added dropwise. The mixture was allowed to warm 0° C. and stirring continued at 0° C. for one hour. A pyridine 4-carboxaldehyde was added to the solution and the reaction was stirred at 0° C. for one hour. The reaction mixture was poured into ethyl acetate and washed with water. The organic layer was separated and dried over MgSO4. Removal of the solvent followed by chromatography on silica gel (hexane:ethyl acetate 2:1 as eluent) afforded 1 g of the desired product.
  • The following example was prepared using Procedure K:
  • Example 41 N-[4-Chloro-2-(hydroxy-pyridin-4-yl-methyl)-phenyl]-2,2-dimethyl-propionamide
  • Figure US20100324093A1-20101223-C00068
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 319. (M+H+).
  • Procedure L
  • The alcohol from Procedure K (1 g) was dissolved in 25 mL of dichloromethane. Manganese dioxide (2 g) was added and the mixture was heated to reflux for 2 hours. After cooling and filtration, the solvent was evaporated to give 0.95 g of the desired ketone.
  • The following example was prepared using Procedure L:
  • Example 42 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-2,2-dimethyl-propionamide
  • Figure US20100324093A1-20101223-C00069
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 317 (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ10.06 (s, 1H), δ 8.87 (d, 2H), δ 8.8 (d, 1H), δ 7.61 (d, 1H), δ 7.50 (d, 2H), δ 7.47 (s, 1H), δ 1.35 (s, 9H).
  • Procedure M
  • A solution of the pivaloyl protected (2-amino-5-chloro-phenyl)-pyridin-4-yl-methanone from Procedure L (2 g) was dissolved in 25 mL of ethanol along with 8 mL of 6 N HCl. The mixture was brought to reflux for 7 hours. The ethanol was removed by rotary evaporation and the solution neutralized with 6 N NaOH. The precipitated solid was collected by filtration, washed with water, and dried to yield (2-amino-5-chloro-phenyl)-pyridin-4-yl-methanone.
  • The following example was prepared using Procedure M:
  • Example 43 (2-Amino-5-chloro-phenyl)-pyridin-4-yl-methanone
  • Figure US20100324093A1-20101223-C00070
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 233. (M+H+). 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 8.80 (d, 2H), δ 7.45 (d, 2H), δ 7.29 (m, 2H), δ 6.70 (d, 1H), δ 6.33 (br s, 2H).
  • (2-Amino-5-chloro-phenyl)-pyridin-4-yl-methanones prepared using the method depicted in Scheme IV can be converted to the corresponding aryl sulfonamide or heteroaryl sulfonamide using Procedure G or H. The following example was prepared using the procedure depicted in Scheme IV followed by Procedure G or H:
  • Example 44 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethyl-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00071
  • LC-MS showed the product to be >95% pure and to have the expected M.W. of 401 (M+H+); 1H NMR (Bruker 300 MHz, CDCl3, shifts relative to the solvent peak at 7.24 ppm) δ 10.0 (s, 1H), δ 7.78 (d, 1H), δ 7.64-7.58 (m, 3H), δ 7.52 (d, 1H) δ 7.34-7.21 (m, 3H), δ 7.17-7.21 (m, 3H), δ 2.58 (m, 2H), δ 1.15 (m, 3H).
  • VI. Synthesis of Sulfones and Sulfoxides
  • Figure US20100324093A1-20101223-C00072
  • Procedure N Production of 4-Chloro-1-nitro-2-phenylsulfanyl-benzene
  • To a solution of 2 g of 2-fluoro-4-chloro-nitrobenzene in 50 ml DMF and 1.25 ml of thiophenol was added 1.9 ml triethylamine at room temperature. The mixture was stirred for one hour. One hundred mL of water was added and a yellow solid crashed out of solution. The solid was collected by filtration and dried. Yield: 50-70%. LC-MS showed a single peak with the expected (M+H+) of 267.
  • Procedure O Production of 4-Chloro-2-phenylsulfanyl-phenylamine
  • To a suspension of 1.8 g of 2-benzylthio-4-chloro-nitrobenzene in 100 ml ethanol was added 2 g powdered iron and 20 mL of 0.33N NH4Cl. The mixture was heated at 70° C. for 6 hr. The mixture was cooled and filtered through a pad of Celite. The filtrate was condensed and purified by flash chromatography (hexane/ethyl acetate 4:1 as eluent) to yield 1.25 g of an oil. LC-MS showed a single peak with the expected (M+H+) of 236.
  • The following example was prepared by Procedure C:
  • Example 116 (4-Chloro-2-phenylsulfanyl-phenyl)-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00073
  • The product showed a single peak by LC-MS with the expected (M+H+) of 434.
  • Procedure P
  • To a solution of the 0.5 g of the sulfide from procedure C in 20 ml DCM, was added one equivalent of m-chloro peroxybenzoic acid (mCPBA). The mixture was stirred at room temperature overnight. The mixture was concentrated and purified by flash chromatography (hexane/ethyl acetate=2:1 as eluent) to give 0.12 g of white solid as product.
  • The following example was prepared using Procedure P:
  • Example 63 N-(2-Benzenesulfinyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00074
  • LC-MS showed a single peak with the expected (M+H+) 450.
  • Procedure Q
  • To a solution of the 0.5 sulfide from procedure C in 20 ml DCM, added two equivalents of mCPBA. The mixture was stirred at room temperature overnight. The mixture is concentrated and purified by flash chromatography (hexane/ethyl acetate=2:1 as eluent) to give 0.14 g of white solid as product.
  • The following example was prepared using Procedure Q.
  • Example 117 N-(2-Benzenesulfonyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00075
  • LC-MS showed a single peak with the expected (M+H+) 466.
  • The following example was prepared using Procedure G:
  • Example 118 N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00076
  • LC-MS showed a single peak with the expected (M+H+) of 431.
  • Figure US20100324093A1-20101223-C00077
  • Procedure R
  • N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzensulfonamide (60 mg) was dissolved in 5 mL of dichloromethane. 1.05 eq. of mCPBA was added and the mixture stirred at room temperature overnight. The solvent was removed by rotary evaporation and the residue chromatographed on silica gel (hexane/ethyl acetate=1:1 as eluent) to give the product. Yield: 95%.
  • The following example was prepared using Procedure R:
  • Example 119 N-[4-Chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide
  • Figure US20100324093A1-20101223-C00078
  • LC-MS showed a single peak with the expected (M+H+) of 447.
  • Other examples listed herein can be made by the general procedures outlined above.
  • Biological Data
  • I. FMAT Assay: FMAT Assay determine inhibitors of TECK binding to human CCR9 receptors:
  • A CCR9 assay buffer was prepared by adding 5 mL of 1M HEPES, 1.25 mL of 1M CaCl2, 1.5 mL of 1M MgCl2, 14 MgCl2, mL of 5M NaCl, 0.5 g fatty acid free bovine serum albumin, and 1 mL of 5% azide were added to 400 mL of distilled, deionized water (ddH2O). The solution was mixed until all the fatty acid free bovine serum albumin was dissolved. After pH was adjusted to 7.4, the final volume was adjusted to 500 mL, and the solution was filtered through a 0.2 μm filter.
  • A 1000 nM biotinylated human TECK in assay buffer was prepared. A 14 μM solution of Cy5-α-Biotin working solution was prepared. Biotinylated TECK and Cy5-α-Biotin working solutions were mixed right before cells were ready to be added into wells. 13 mL of assay buffer was added to a 50 mL polyethylene tube. 5.6 μL of 1000 nM biotinylated TECK working solution and 178 μL of 14 μg/mL Cy5-α-Biotin working solution were added to the tube and mixed. The final biotinylated TECK concentration was 0.4 nM and the final Cy5-α-Biotin concentration 0.064 μg/mL.
  • Cells were plated right before all other reagents are added to wells. Cells were collected and centrifuged in a tabletop centrifuge at 1200 rpm for 5 minutes. The supernatant was removed by vacuum without disturbing cell pellet. The cells were resuspended in 5 mL of assay buffer. The cells were counted by adding 1 mL of a cell suspension to an eppendorf tube. Then 150 μL of this suspension was added to 150 μL of Trypan Blue. Then 10 μL of Trypan Blue suspension was added to hemocytometer, and the cell number per mL was calculated. The cells were resuspended with CCR9 assay buffer to final 0.2×106 cells/mL for the assay. Approximately 2.6×106 cells for 1 384 well plate was needed.
  • 20 μL of a test compound dissolved in DMSO was added to a well a polypropylene 384 well plate. Multiple compounds were tested simultaneously by adding a different test compound to each well. A control of 20 μL of DMSO in assay buffer was prepared. 20 μL of the Biotin-Teck and Cy5-α-Biotin mixed solution was added to each well of the polypropylene plate. 20 μL of cells suspension at 0.2×106/mL was added to each well. The assay plate was incubated on a rotator at room temperature for 1.5 hour. FMAT was read (i 490/518 or 537/568, threshold set at 1 standard deviation).
  • II. FLIPR Assay: FLIPR Assay determines inhibitors of TECK induced response in Chinese Hamster Ovary (CHO) cells that over express recombinant human CCR9 and the Ga16 protein.
  • The day before the assay was run, CHO cells were diluted to give appropriately 10,000 cells/well (in a volume of 50 mL). Each well of the 384 black/clear plate was then seeded with 50 mL of the diluted cell suspension. The cell plates were placed in a 37° C. tissue culture incubator at 6% CO2 overnight.
  • A wash buffer and a dye loading solution were prepared on the day the assay was preformed. The wash buffer was prepared by mixing 880 mL of Nanopure water, 100 mL of 10×HBSS and 20 mL of 1M HEPES to give 1 L of a 1×HBSS and 20 mM HEPES solution. 1 g of BSA (bovine serum albumin) was added to the 1×HBSS/20 mM HEPES solution. A 250 mM probenecid stock solution was prepared by dissolving 710 mg of probenecid in 5 mL of 1 N NaOH and 5 mL of previously prepared 1×HBSS/20 mM HEPES/0.1% BSA buffer. 10 mL of 250 mM probenecid was added to the 1×HBSS/20 mM HEPES/0.1% BSA buffer to give a wash buffer having 1×HBSS/20 mM HEPES/2.5 mM probenecid/0.1% BSA.
  • For each 384 well plate, 11 mL of 2×FLUO-3 dye loading solution was required. The 2×FLUO-3 dye loading solution prepared by adding 22 mL of DMSO (100%) to each of 2×50 mg vials of FLUO-3. The vials were vortexed, then 22 mL of 20% pluronic acid was added to each 50 mg vial of FLUO-3 and vortex. 88 mL of reconstituted FLUO-3 stock solution was added to 11 mL of 1× wash buffer.
  • Each cell plate was washed with the wash buffer. At the end of the wash, there was 25 mL residual volume per well. 25 mL of dye loading solution was added to each well of the 384 plate. The plates were placed in an incubator for at least 1 hour. A yellowplate calibration plate was run on a FLIPR 384, and a standard deviation of less than 3.5% was obtained.
  • 10 mM stock concentrations of compounds in 100% DMSO were prepared and stored at room temperature. Stock concentrations (100× final assay concentration) 9×1:3 serial dilutions in 100% DMSO were prepared in order to carry out 10 point concentration response curves. 3× final assay concentration were prepared by making 1 in 33.3 dilutions of the 100× stocks in wash buffer and were plated into Greiner 384 well plates. 10 point concentration response curves started at 100 mM, 10 mM or 1 mM (final assay concentration). The 1st compound was added to wells A1-A10 and other compounds were added down the plate to P1-P10. Columns 11 and 12 were control wells, with FLIPR buffer (with 3% DMSO) added to wells A11-H12. The remaining compounds were added to A13-A22 down through P13-P22. Each compound was tested in triplicate.
  • Ligand (rhTECK) plate at 4× ligand EC50 (final assay concentration) in wash buffer was prepared and plated into a Greiner 384 well plate. Appropriate amount of 4× ligand was added to wells A1 through P10 and A13 through P22. In columns 11 and 12, FLIPR wash buffer was added to wells A11-D12 and 4× ligand solution was added to wells E11-H12.
  • The cell plates were incubated at least a 1 hour with the dye solution, then washed with wash buffer. At the end of the wash, there was 25 mL residual volume per well. The cell plate were loaded onto the stacker of FLIPR-384. Individual well activity (IWA) was measured, using the max-min function between timepoints 88-145. Data was expressed as % inhibition of rhTECK induced response, and IC50 values were calculated for compounds displaying antagonist activity.
  • Inhibition of binding of human TECK to human CCR9 receptors (FMAT assay) and inhibition of human TECK induced response(FLIPR)
  • Of the compounds tested in the FMAT assay described above, the following provided an IC50 value less than or equal to about 1.0 μM: 17-21, 31-34, 44, 70, 73, 74, and 115.
  • Of the compounds tested in the FLIPR assay described above, the following provided an IC50 value less than or equal to about 1.0 μM: 8-13, 14-15, 17-20, 22-24, 31-36, 44, 49, 63, and 69-89, 116-143.

Claims (22)

1. A compound represented by the following structural formula:
Figure US20100324093A1-20101223-C00079
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
Y is C(O), O, S, S(O), or S(O)2;
X1, X2, and X3 are each, independently, N or CR, provided that at least one of
X1, X2, or X3 is CR;
R for each occurrence and R1 are each, independently, H or a substituent;
R6 is H, an aliphatic carbonyl group, or an aliphatic ester;
ring A is substituted or unsubstituted; and
Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl or a substituted or unsubstituted heteroaryl.
2. The compound of claim 1, wherein Ar2 is phenyl, naphthyl, thienyl, or thianaphthenyl, each group being substituted or unsubstituted.
3. The compound of claim 1, wherein Ar1 is phenyl, pyridyl, pyrimidinyl, or pyrazinyl, each group being substituted or unsubstituted.
4. The compound of claim 1, which is represented by the following structural formula:
Figure US20100324093A1-20101223-C00080
or a pharmaceutically acceptable salts, solvates, or hydrates thereof,
wherein: Y is S, O, S(O), or S(O)2;
R19 and R20 are each, independently, H or a substituent.
5. The compound of claim 1, which is represented by the following structural formula:
Figure US20100324093A1-20101223-C00081
or pharmaceutically acceptable salts, solvates, or hydrates thereof,
wherein: R1, R2, R3, R4, R5, and each R are, independently, H, an aliphatic group, haloalkyl, halo, COOH, NO2, alkoxy, or haloalkoxy; and
X4 is CR, N or N+—O.
6. The compound of claim 1 which is represented by the following structural formula:
Figure US20100324093A1-20101223-C00082
or pharmaceutically acceptable salts, solvates, or hydrates thereof,
wherein:
X4 is CR, N or N+—O;
R8 is H or an electron withdrawing group;
m and n are each, independently, 0 or an integer from 1 to 3;
each R9 is, independently, aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12; or two adjacent R9 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R10 is, independently, halo, an aliphatic group, alkoxy, or haloalkyl; or two adjacent R10 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R11 is, independently, H or an aliphatic group; and
R12 is an aliphatic group.
7. The compound of claim 1 wherein the compound is represented by the following structural formula:
Figure US20100324093A1-20101223-C00083
or pharmaceutically acceptable salts, solvates, or hydrates thereof,
wherein:
ring B is substituted or unsubstituted;
p is 0 or an integer from 1-3; and
each R13 is, independently, a halo or a substituted or unsubstituted heteroaryl.
8. A compound selected from the group consisting of:
N-[4-Chloro-2-(2-chloro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide Thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(2-Benzoyl-4-chloro-phenyl)-4-iodo-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-chloro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-tert-butyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-propyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-isopropyl-benzenesulfonamide 4-Ethyl-N-[4-nitro-2-(3-trifluoromethyl-benzoyl)-phenyl]-benzenesulfonamide N-[4-Chloro-2-(3-methyl-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-chloro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-propoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-butoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-benzyloxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-phenoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropyl-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isobutyl-benzenesulfonamide N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-ethoxy-benzenesulfonamide N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-trifluoromethyl-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-chloro-benzenesulfonamide 5-Benzoyl-6-(4-isopropoxy-benzenesulfonylamino)-nicotinic acid N-(2-Benzoyl-4-chloro-phenyl)-2-nitro-benzenesulfonamide N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(5-Benzoyl-pyrimidin-4-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-trifluoromethoxy-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(5-Benzoyl-2-trifluoromethyl-pyrimidin-4-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclobutyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-tert-butoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopentyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylsulfanyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylamino-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclohexyl-benzenesulfonamide N-(3-Benzenesulfinyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzenesulfonyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridazine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-trifluoromethoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-methoxy-benzenesulfonamide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethylamino-benzenesulfonamide 5-(1-Methyl-5-trifluoromethyl-1H-pyrazol-3-yl)-thiophene-2-sulfonic acid (2-benzoyl-4- chloro-phenyl)-amide N-(2-Benzoyl-4-iodo-phenyl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(3-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-nitro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-diethylamino-benzenesulfonamide N-(2-Benzoyl-4-fluoro-phenyl)-4-methoxy-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(pyridine-3-carbonyl)-phenyl]-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-methyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-bromo-benzenesulfonamide N-[4-Chloro-2-(pyridine-3-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-dimethylamino-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-fluoro-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(2-fluoro-benzoyl)-phenyl]-benzenesulfonamide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide Thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide 5-Bromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide 5-Chloro-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(pyridine-2-carbonyl)-phenyl]-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-benzenesulfonamide N-[4-(2-Benzoyl-4-chloro-phenylsulfamoyl)-phenyl]-acetamide N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-cyano-benzenesulfonamide 4,5-Dibromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(pyridine-3-sulfonyl)-phenyl]-4-nitro-benzenesulfonamide Naphthalene-2-sulfonic acid [4-chloro-2-(pyridine-3-carbonyl)-phenyl]-amide N-(2-Benzoyl-4-chloro-phenyl)-3-bromo-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-methyl-3-nitro-benzenesulfonamide N-[4-Chloro-2-(2-methoxy-benzoyl)-phenyl]-4-nitro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-2-trifluoromethyl-benzenesulfonamide 5-Chloro-3-methyl-benzo[b]thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)- amide N-(2-Benzoyl-phenyl)-4-nitro-benzenesulfonamide 5-Isoxazol-3-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(2-Benzoyl-phenyl)-4-methyl-benzenesulfonamide Benzo[b]thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(4-Chloro-2-phenylsulfanyl-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzenesulfonyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide 4-Isopropoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]- benzenesulfonamide 4-Ethoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]-benzenesulfonamide N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropyl-benzenesulfonamide N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-[5-Chloro-3-(thiophene-2-carbonyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid [5-chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]- amide N-[3-(Benzofuran-2-carbonyl)-5-chloro-pyridin-2-yl]-4-isopropoxy- benzenesulfonamide N-(2-Benzoyl-4-trifluoromethyl-phenyl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-pyridin-4-yl)-4-isopropoxy-benzenes N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropyl-benzenesulfonamide N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropylamino-benzenesulfonamide N-(4-Benzoyl-pyridin-3-yl)-4-isopropyl-benzenesulfonamide N-(4-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide N-[3-(3-Fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropoxy-benzenesulfonamide 4-Isopropoxy-N-[3-(pyridine-4-carbonyl)-pyridin-4-yl]-benzenesulfonamide 6-Isopropoxy-pyridine-3-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide 6-Isopropoxy-pyridine-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(pyridin-2-ylsulfanyl)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-phenyl)-4-isopropoxy-benzenesulfonamide; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
9. A pharmaceutical composition, comprising a pharmaceutically acceptable carrier and the compound of claim 1.
10. A method of inhibiting a CCR9 receptor function in a subject in need thereof, comprising the step of administering to the subject an effective amount of a compound represented by the following structural formula:
Figure US20100324093A1-20101223-C00084
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
Y is C(O), S, O, S(O) or S(O)2;
X1, X2, and X3 are each, independently, N or CR, provided that at least one of X1, X2, or X3 is CR;
R, for each occurrence, and R1 are each, independently, H or a substituent;
R6 is H, an aliphatic carbonyl group, or an aliphatic ester; and
ring A is substituted or unsubstituted;
Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group.
11. The method of claim 10, wherein the compound is represented by a structural formula selected from the group consisting of A, B, C, and D:
A.
Figure US20100324093A1-20101223-C00085
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R19 and R20 are each, independently, H or a substituent;
B.
Figure US20100324093A1-20101223-C00086
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R1, R2, R3, R4, R5, and each R are, independently, H, an aliphatic group, haloalkyl, a halo, COOH, NO2, alkoxy, or haloalkoxy; and
X4 is CR, N or N+—O;
C.
Figure US20100324093A1-20101223-C00087
or pharmaceutically acceptable salts, solvates or hydrates thereof, wherein:
X4 is CR, N or N+—O;
R8 is H or an electron withdrawing group;
m and n are each, independently, 0 or an integer from 1 to 3;
each R9 is, independently, an aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12; or two adjacent R9 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R10 is, independently, halo, aliphatic group, alkoxy, or haloalkyl; or two adjacent R10 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R11 is, independently, H or an aliphatic group; and
R12 is an aliphatic group; and
D.
Figure US20100324093A1-20101223-C00088
or pharmaceutically acceptable salts, solvates or hydrates thereof, wherein:
R8 is halo, nitro, alkylcarbonyl or trihaloalkyl;
p is 0 or an integer from 1 to 3; and
each R13 is, independently, a halo, a substituted or unsubstituted heterocycle, or a substituted or unsubstituted heteroaryl.
12. The method of claim 10 wherein the subject is being treated for an inflammatory disease or condition.
13. A method of treating or preventing an inflammatory disease or condition in a subject in need thereof, comprising administering to the subject an effective amount of a compound represented by the following structural formula:
Figure US20100324093A1-20101223-C00089
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
Y is C(O), S, O, S(O) or S(O)2;
X1, X2, and X3 are each, independently, N or CR, provided that at least one of X1, X2, or X3 is CR;
R, for each occurrence, and R1 are each, independently, H or a substituent; and
ring A is substituted or unsubstituted;
R6 is H, an aliphatic group, an aryl group or an aliphatic carbonyl group; and
Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group.
14. The method of claim 13, wherein the compound is represented by a structural formula selected from the group consisting of A, B, C, and D:
Figure US20100324093A1-20101223-C00090
A.
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R19 and R20 are each, independently, H or a substituent.
B.
Figure US20100324093A1-20101223-C00091
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R1, R2, R3, R4, R5, and each R are, independently, H, an aliphatic group, haloalkyl, halo, COOH, NO2, alkoxy, or haloalkoxy; and
X4 is CR, N or N+—O;
C.
Figure US20100324093A1-20101223-C00092
or pharmaceutically acceptable salts, solvates or hydrates thereof, wherein:
X4 is CR, N or N+—O;
R8 is H or an electron withdrawing group;
m and n are each, independently, 0 or an integer from 1 to 3;
each R9 is, independently, aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12; or two adjacent R9 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R10 is, independently, halo, aliphatic group, alkoxy, or haloalkyl; or two adjacent R10 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R11 is, independently, H or an aliphatic group; and
R12 is an aliphatic group; and
Figure US20100324093A1-20101223-C00093
D.
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
R8 is a halo, nitro, alkylcarbonyl or trihaloalkyl;
p is 0 or an integer from 1 to 3; and
each R13 is, independently, a halo, a substituted or unsubstituted heterocycle, or a substituted or unsubstituted heteroaryl.
15. The method of claim 13, wherein the compound is selected from the group consisting of:
N-[4-Chloro-2-(2-chloro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide Thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(2-Benzoyl-4-chloro-phenyl)-4-iodo-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-chloro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-tert-butyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-propyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-isopropyl-benzenesulfonamide 4-Ethyl-N-[4-nitro-2-(3-trifluoromethyl-benzoyl)-phenyl]-benzenesulfonamide N-[4-Chloro-2-(3-methyl-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-chloro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-propoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-butoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-benzyloxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-phenoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-ethoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropyl-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isobutyl-benzenesulfonamide N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-ethoxy-benzenesulfonamide N-[5-Chloro-3-(pyridine-4-carbonyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-trifluoromethyl-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-chloro-benzenesulfonamide 5-Benzoyl-6-(4-isopropoxy-benzenesulfonylamino)-nicotinic acid N-(2-Benzoyl-4-chloro-phenyl)-2-nitro-benzenesulfonamide N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(5-Benzoyl-pyrimidin-4-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-trifluoromethoxy-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(5-Benzoyl-2-trifluoromethyl-pyrimidin-4-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclobutyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-tert-butoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopentyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylsulfanyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropylamino-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclopropyl-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-cyclohexyl-benzenesulfonamide N-(3-Benzenesulfinyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzenesulfonyl-5-chloro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridazine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(2-trifluoromethyl-pyrimidine-5-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-[4-Chloro-2-(6-trifluoromethyl-pyridazine-4-carbonyl)-phenyl]-4-isopropoxy- benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzoyl-4-nitro-phenyl)-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-trifluoromethoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-methoxy-benzenesulfonamide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-ethyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-ethylamino-benzenesulfonamide 5-(1-Methyl-5-trifluoromethyl-1H-pyrazol-3-yl)-thiophene-2-sulfonic acid (2-benzoyl-4- chloro-phenyl)-amide N-(2-Benzoyl-4-iodo-phenyl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(3-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide N-[4-Chloro-2-(3-chloro-benzoyl)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-nitro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-diethylamino-benzenesulfonamide N-(2-Benzoyl-4-fluoro-phenyl)-4-methoxy-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(pyridine-3-carbonyl)-phenyl]-benzenesulfonamide N-(2-Benzoyl-4-bromo-phenyl)-4-methyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-bromo-benzenesulfonamide N-[4-Chloro-2-(pyridine-3-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-dimethylamino-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-fluoro-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(2-fluoro-benzoyl)-phenyl]-benzenesulfonamide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methoxy-benzenesulfonamide Thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide 5-Bromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide 5-Chloro-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-nitro-benzenesulfonamide N-[4-Chloro-2-(2-fluoro-benzoyl)-phenyl]-4-methyl-benzenesulfonamide 4-Chloro-N-[4-chloro-2-(pyridine-2-carbonyl)-phenyl]-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-benzenesulfonamide N-[4-(2-Benzoyl-4-chloro-phenylsulfamoyl)-phenyl]-acetamide N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-2-carbonyl)-phenyl]-4-methyl-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-cyano-benzenesulfonamide 4,5-Dibromo-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(pyridine-3-sulfonyl)-phenyl]-4-nitro-benzenesulfonamide Naphthalene-2-sulfonic acid [4-chloro-2-(pyridine-3-carbonyl)-phenyl]-amide N-(2-Benzoyl-4-chloro-phenyl)-3-bromo-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-4-methyl-3-nitro-benzenesulfonamide N-[4-Chloro-2-(2-methoxy-benzoyl)-phenyl]-4-nitro-benzenesulfonamide N-(2-Benzoyl-4-chloro-phenyl)-2-trifluoromethyl-benzenesulfonamide 5-Chloro-3-methyl-benzo[b]thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)- amide N-(2-Benzoyl-phenyl)-4-nitro-benzenesulfonamide 5-Isoxazol-3-yl-thiophene-2-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(2-Benzoyl-phenyl)-4-methyl-benzenesulfonamide Benzo[b]thiophene-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-(4-Chloro-2-phenylsulfanyl-phenyl)-4-isopropoxy-benzenesulfonamide N-(2-Benzenesulfonyl-4-chloro-phenyl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-4-isopropoxy-benzenesulfonamide 4-Isopropoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]- benzenesulfonamide 4-Ethoxy-N-[2-(pyridine-4-carbonyl)-4-trifluoromethyl-phenyl]-benzenesulfonamide N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropyl-benzenesulfonamide N-[5-Chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-[5-Chloro-3-(thiophene-2-carbonyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid [5-chloro-3-(3-fluoro-benzoyl)-pyridin-2-yl]- amide N-[3-(Benzofuran-2-carbonyl)-5-chloro-pyridin-2-yl]-4-isopropoxy- benzenesulfonamide N-(2-Benzoyl-4-trifluoromethyl-phenyl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-pyridin-4-yl)-4-isopropoxy-benzenesulfonamide N-(3-Benzoyl-5-chloro-pyridin-2-yl)-4-isopropyl-benzenesulfonamide N-(3-Benzoyl-5-nitro-pyridin-2-yl)-4-isopropoxy-benzenesulfonamide 5-Oxazol-5-yl-thiophene-2-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide N-(2-Benzoyl-4-chloro-phenyl)-4-isopropylamino-benzenesulfonamide N-(4-Benzoyl-pyridin-3-yl)-4-isopropyl-benzenesulfonamide N-(4-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-pyridin-3-yl)-4-isopropoxy-benzenesulfonamide N-[3-(3-Fluoro-benzoyl)-pyridin-2-yl]-4-isopropoxy-benzenesulfonamide N-[4-Chloro-2-(pyridin-3-yloxy)-phenyl]-4-isopropoxy-benzenesulfonamide 4-Isopropoxy-N-[3-(pyridine-4-carbonyl)-pyridin-4-yl]-benzenesulfonamide 6-Isopropoxy-pyridine-3-sulfonic acid (3-benzoyl-5-chloro-pyridin-2-yl)-amide 6-Isopropoxy-pyridine-3-sulfonic acid (2-benzoyl-4-chloro-phenyl)-amide N-[4-Chloro-2-(pyridin-2-ylsulfanyl)-phenyl]-4-isopropyl-benzenesulfonamide N-(2-Benzoyl-phenyl)-4-isopropoxy-benzenesulfonamide; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
16. The method of claim 13, wherein the inflammatory disease or condition is Crohn's disease or colitis.
17. The method of claim 13, wherein the inflammatory disease or condition is Celiac's disease.
18. The method of claim 13, wherein the compound inhibits the binding of a ligand to CCR9.
19. The method of claim 18, wherein the ligand is TECK.
20. A method of inhibiting CCR9-mediated homing of leukocytes in a subject in need of such treatment comprising administering to the subject an effective amount of at least one compound represented by the following structural formula:
Figure US20100324093A1-20101223-C00094
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
Y is C(O), S, S(O) or S(O)2;
X1, X2, and X3 are each, independently, N or CR, provided that at least one of X1, X2, or X3 is CR;
R, for each occurrence, and R1 are each, independently, H or a substituent;
R6 is H, an aliphatic group, an aryl group or an aliphatic carbonyl group;
ring A is substituted or unsubstituted; and
Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group.
21. The method of claim 20, wherein the compound is represented by a structural formula selected from the group consisting of A, B, C, and D:
A.
Figure US20100324093A1-20101223-C00095
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R19 and R20 are each, independently, H or a substituent.
B.
Figure US20100324093A1-20101223-C00096
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein: R1, R2, R3, R4, R5, and each R are, independently, H, an aliphatic group, haloalkyl, halo, COOH, NO2, alkoxy, or haloalkoxy; and
X4 is CR, N or N+—O−;
C.
Figure US20100324093A1-20101223-C00097
or pharmaceutically acceptable salts, solvates or hydrates thereof, wherein:
X4 is CR, N or N+—O;
R8 is H or an electron withdrawing group;
m and n are each, independently, 0 or an integer from 1 to 3;
each R9 is, independently, an aliphatic group, haloalkyl, aryl, arylalkyl, alkoxy, cycloalkoxy, haloalkoxy, aryloxy, arylalkoxy, alkylthio, halo, nitro, cyano, hydroxy, NR11CO2R12, C(O)N(R11)2, C(O)R12, CO2R12, OC(O)N(R11)2, OC(O)R12, N(R11)2, or NR11C(O)R12; or two adjacent R9 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R10 is, independently, halo, aliphatic group, alkoxy, or haloalkyl; or two adjacent R10 groups taken together with the atoms to which they are attached form a fused, saturated, unsaturated or partially unsaturated 5 to 7 membered ring having 0, 1 or 2 heteroatoms selected from the group consisting of N, O, and S;
each R11 is, independently, H or an aliphatic group; and
R12 is an aliphatic group; and
D.
Figure US20100324093A1-20101223-C00098
or pharmaceutically acceptable salts, solvates or hydrates thereof,
wherein:
R8 is a halo, nitro, alkylcarbonyl or trihaloalkyl;
p is 0 or an integer from 1 to 3; and
each R13 is, independently, a halo, a substituted or unsubstituted heterocycle, or a substituted or unsubstituted heteroaryl.
22. A method of inhibiting homing of leukocytes to mucosal tissue in a subject in need of such treatment, comprising administering to the subject an effective amount of at least one compound represented by the following structural formula:
Figure US20100324093A1-20101223-C00099
or pharmaceutically acceptable salts, solvates or hydrates thereof, wherein:
Y is C(O), S, S(O) or S(O)2;
X1, X2, and X3 are each, independently, N or CR, provided that at least one of X1, X2, or X3 is CR;
R, for each occurrence, and R1 are each, independently, H or a substituent;
R6 is H, an aliphatic group, an aryl group or an aliphatic carbonyl group;
ring A is substituted or unsubstituted; and
Ar1 and Ar2 are each, independently, a substituted or unsubstituted aryl group or a substituted or unsubstituted heteroaryl group.
US12/869,070 2002-05-24 2010-08-26 Ccr9 inhibitors and methods of use thereof Abandoned US20100324093A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/869,070 US20100324093A1 (en) 2002-05-24 2010-08-26 Ccr9 inhibitors and methods of use thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US38357302P 2002-05-24 2002-05-24
US10/443,155 US7238717B2 (en) 2002-05-24 2003-05-21 CCR9 inhibitors and methods of use thereof
US11/391,633 US7282502B2 (en) 2002-05-24 2006-03-28 CCR9 inhibitors and methods of use thereof
US11/974,850 US7659272B2 (en) 2002-05-24 2007-10-16 CCR9 inhibitors and methods of use thereof
US12/640,439 US7999109B2 (en) 2002-05-24 2009-12-17 CCR9 inhibitors and methods of use thereof
US12/869,070 US20100324093A1 (en) 2002-05-24 2010-08-26 Ccr9 inhibitors and methods of use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/640,439 Continuation US7999109B2 (en) 2002-05-24 2009-12-17 CCR9 inhibitors and methods of use thereof

Publications (1)

Publication Number Publication Date
US20100324093A1 true US20100324093A1 (en) 2010-12-23

Family

ID=29584585

Family Applications (11)

Application Number Title Priority Date Filing Date
US10/443,155 Expired - Lifetime US7238717B2 (en) 2002-05-24 2003-05-21 CCR9 inhibitors and methods of use thereof
US11/391,633 Expired - Lifetime US7282502B2 (en) 2002-05-24 2006-03-28 CCR9 inhibitors and methods of use thereof
US11/601,025 Abandoned US20070066823A1 (en) 2002-05-24 2006-11-17 CCR9 inhibitors and methods of use thereof
US11/974,850 Expired - Fee Related US7659272B2 (en) 2002-05-24 2007-10-16 CCR9 inhibitors and methods of use thereof
US12/288,075 Expired - Fee Related US7820717B2 (en) 2002-05-24 2008-10-16 CCR9 inhibitors and methods of use thereof
US12/630,040 Expired - Fee Related US8030517B2 (en) 2002-05-24 2009-12-03 CCR9 inhibitors and methods of use thereof
US12/640,439 Expired - Fee Related US7999109B2 (en) 2002-05-24 2009-12-17 CCR9 inhibitors and methods of use thereof
US12/869,070 Abandoned US20100324093A1 (en) 2002-05-24 2010-08-26 Ccr9 inhibitors and methods of use thereof
US13/221,315 Expired - Fee Related US8569543B2 (en) 2002-05-24 2011-08-30 CCR9 inhibitors and methods of use thereof
US14/049,452 Expired - Lifetime US9035096B2 (en) 2002-05-24 2013-10-09 CCR9 inhibitors and methods of use thereof
US14/687,303 Abandoned US20160068479A1 (en) 2002-05-24 2015-04-15 Ccr9 inhibitors and methods of use thereof

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US10/443,155 Expired - Lifetime US7238717B2 (en) 2002-05-24 2003-05-21 CCR9 inhibitors and methods of use thereof
US11/391,633 Expired - Lifetime US7282502B2 (en) 2002-05-24 2006-03-28 CCR9 inhibitors and methods of use thereof
US11/601,025 Abandoned US20070066823A1 (en) 2002-05-24 2006-11-17 CCR9 inhibitors and methods of use thereof
US11/974,850 Expired - Fee Related US7659272B2 (en) 2002-05-24 2007-10-16 CCR9 inhibitors and methods of use thereof
US12/288,075 Expired - Fee Related US7820717B2 (en) 2002-05-24 2008-10-16 CCR9 inhibitors and methods of use thereof
US12/630,040 Expired - Fee Related US8030517B2 (en) 2002-05-24 2009-12-03 CCR9 inhibitors and methods of use thereof
US12/640,439 Expired - Fee Related US7999109B2 (en) 2002-05-24 2009-12-17 CCR9 inhibitors and methods of use thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/221,315 Expired - Fee Related US8569543B2 (en) 2002-05-24 2011-08-30 CCR9 inhibitors and methods of use thereof
US14/049,452 Expired - Lifetime US9035096B2 (en) 2002-05-24 2013-10-09 CCR9 inhibitors and methods of use thereof
US14/687,303 Abandoned US20160068479A1 (en) 2002-05-24 2015-04-15 Ccr9 inhibitors and methods of use thereof

Country Status (9)

Country Link
US (11) US7238717B2 (en)
EP (4) EP2399903A1 (en)
JP (7) JP4828822B2 (en)
AU (2) AU2003248549B2 (en)
CA (2) CA2485681C (en)
MX (1) MXPA04011465A (en)
NZ (2) NZ566263A (en)
WO (1) WO2003099773A1 (en)
ZA (1) ZA200409131B (en)

Families Citing this family (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003229767A1 (en) * 2002-05-14 2003-11-11 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with chemokine receptor 9(ccr9)
AU2003248549B2 (en) * 2002-05-24 2010-04-08 Millennium Pharmaceuticals, Inc. CCR9 inhibitors and methods of use thereof
US7420055B2 (en) * 2002-11-18 2008-09-02 Chemocentryx, Inc. Aryl sulfonamides
US7227035B2 (en) * 2002-11-18 2007-06-05 Chemocentryx Bis-aryl sulfonamides
US7741519B2 (en) 2007-04-23 2010-06-22 Chemocentryx, Inc. Bis-aryl sulfonamides
EP1562940B1 (en) * 2002-11-18 2007-05-30 ChemoCentryx Inc Aryl sulfonamides
AU2004212985B2 (en) 2003-02-20 2010-10-14 Encysive Pharmaceuticals Inc. Phenylenediamine urotensin-II receptor antagonists and CCR-9 antagonists
US7288538B2 (en) 2003-02-20 2007-10-30 Encysive Pharmaceuticals, Inc. Phenylenediamine urotensin-II receptor antagonists and CCR-9 antagonists
EP1603884A4 (en) 2003-02-28 2008-05-28 Encysive Pharmaceuticals Inc Pyridine, pyrimidine, quinoline, quinazoline, and naphthalene urotensin-ii receptor antagonists.
US7320989B2 (en) 2003-02-28 2008-01-22 Encysive Pharmaceuticals, Inc. Pyridine, pyrimidine, quinoline, quinazoline, and naphthalene urotensin-II receptor antagonists
DE102004027119A1 (en) 2003-06-06 2004-12-30 Schott Ag Production of a UV-absorbed glass used in the production of gas discharge lamps, fluorescent lamps, xenon lamps, LCD displays, computer monitors and telephone displays comprises melting a raw material and producing a melt
US7393873B2 (en) 2003-07-02 2008-07-01 Merck & Co., Inc. Arylsulfonamide derivatives
KR20060101772A (en) * 2003-12-19 2006-09-26 화이자 인코포레이티드 Benzenesulfonylamino-pyridin-2-yl derivatives and related compounds as inhibitors of 11-beta-hydroxysteroid dehydrogenase type 1 (11-beta-hsd-1) for the treatment of diabetes and obesity
US7381738B2 (en) 2004-02-19 2008-06-03 Bristol-Myers Squibb Company Substituted bicycloalkylamine derivatives as modulators of chemokine receptor activity
US7230022B2 (en) 2004-02-19 2007-06-12 Bristol-Myers Squibb Company Substituted fused bicyclic amines as modulators of chemokine receptor activity
US7479496B2 (en) 2004-02-19 2009-01-20 Bristol-Myers Squibb Company Substituted spiro azabicyclics as modulators of chemokine receptor activity
US7288563B2 (en) 2004-02-19 2007-10-30 Bristol-Myers Squibb Company Substituted bicycloalkylamine derivatives as modulators of chemokine receptor activity
JP2007537275A (en) 2004-05-12 2007-12-20 ケモセントリックス, インコーポレイテッド Arylsulfonamide
CN1972918B (en) 2004-06-28 2010-10-13 霍夫曼-拉罗奇有限公司 Pyrimidine derivatives
BRPI0515931A (en) 2004-09-29 2008-08-12 Hoffmann La Roche compounds, process for their preparation, pharmaceutical compositions comprising them, method for treating and / or prophylaxis of diseases and use of such compounds
SE0402635D0 (en) * 2004-10-29 2004-10-29 Astrazeneca Ab Chemical compounds
WO2008109782A2 (en) * 2007-03-06 2008-09-12 Cedars-Sinai Medical Center Diagnosis of inflammatory bowel disease in children
WO2006063093A2 (en) * 2004-12-08 2006-06-15 Cedars-Sinai Medical Center Methods for diagnosis and treatment of crohn's disease
PL2354126T3 (en) * 2005-01-14 2014-03-31 Chemocentryx Inc Heteroaryl sulfonamides and CCR2
US7622583B2 (en) 2005-01-14 2009-11-24 Chemocentryx, Inc. Heteroaryl sulfonamides and CCR2
MX2007016319A (en) 2005-06-16 2008-03-10 Pfizer N-(pyridin-2-yl)-sulfonamide derivatives.
FI20055498A0 (en) * 2005-09-16 2005-09-16 Biotie Therapies Corp Sulfonamides
GB0524786D0 (en) * 2005-12-05 2006-01-11 Glaxo Group Ltd Compounds
GB0526255D0 (en) * 2005-12-22 2006-02-01 Novartis Ag Organic compounds
GB0526252D0 (en) * 2005-12-22 2006-02-01 Novartis Ag Organic compounds
GB0526445D0 (en) * 2005-12-23 2006-02-08 Novartis Ag Organic compounds
US8519135B2 (en) 2006-07-14 2013-08-27 Chemocentryx, Inc. Heteroaryl sulfonamides and CCR2/CCR9
WO2008008374A2 (en) * 2006-07-14 2008-01-17 Chemocentryx, Inc. Ccr2 inhibitors and methods of use thereof
CA2657776C (en) 2006-07-14 2013-08-27 Chemocentryx, Inc. Triazolyl phenyl benzenesulfonamides
HUP0600810A3 (en) * 2006-10-27 2008-09-29 Richter Gedeon Nyrt New sulfonamide derivatives as bradykinin antagonists, process and intermediates for their preparation and pharmaceutical compositions containing them
TW200829578A (en) 2006-11-23 2008-07-16 Astrazeneca Ab Chemical compounds 537
JO2754B1 (en) 2006-12-21 2014-03-15 استرازينكا ايه بي Indazolyl amide derivatives for the treatment of glucocorticoid receptor mediated disorders
JP2010518014A (en) * 2007-01-31 2010-05-27 バーテックス ファーマシューティカルズ インコーポレイテッド 2-Aminopyridine derivatives useful as kinase inhibitors
US20100021917A1 (en) * 2007-02-14 2010-01-28 Cedars-Sinai Medical Center Methods of using genes and genetic variants to predict or diagnose inflammatory bowel disease
WO2008106451A2 (en) * 2007-02-26 2008-09-04 Cedars-Sinai Medical Center Methods of using single nucleotide polymorphisms in the tl1a gene to predict or diagnose inflammatory bowel disease
WO2010039931A2 (en) * 2008-10-01 2010-04-08 Cedars-Sinai Medical Center Methods of using il17rd and il23-il17 pathway genes to diagnose crohn's disease
WO2008116150A2 (en) 2007-03-21 2008-09-25 Cedars-Sinai Medical Center Ileal pouch-anal anastomosis (ipaa) factors in the treatment of inflammatory bowel disease
WO2008134569A2 (en) * 2007-04-26 2008-11-06 Cedars-Sinai Medical Center Diagnosis and treatment of inflammatory bowel disease in the puerto rican population
US20100144903A1 (en) * 2007-05-04 2010-06-10 Cedars-Sinai Medical Center Methods of diagnosis and treatment of crohn's disease
US7776877B2 (en) 2007-06-22 2010-08-17 Chemocentryx, Inc. N-(2-(hetaryl)aryl) arylsulfonamides and N-(2-(hetaryl) hetaryl arylsulfonamides
CN101820881B (en) 2007-07-12 2013-05-01 坎莫森特里克斯公司 Fused heteroaryl pyridyl and phenyl benzenesuflonamides as CCR2 modulators for the treament of inflammation
US20090028866A1 (en) * 2007-07-27 2009-01-29 John Wayne Cancer Institute USE OF CCR9, CCL25/TECK, AND NITEGRIN alpha4 IN DIAGNOSIS AND TREATMENT OF MELANOMA METASTASIS IN THE SMALL INTESTINE
UY31832A (en) 2008-05-20 2010-01-05 Astrazeneca Ab INDAZOL DERIVATIVES REPLACED WITH PHENYL AND BENZODIOXINYL
CN102131807B (en) 2008-07-23 2013-11-20 沃泰克斯药物股份有限公司 Pyrazolopyridine kinase inhibitors
US8569337B2 (en) 2008-07-23 2013-10-29 Vertex Pharmaceuticals Incorporated Tri-cyclic pyrazolopyridine kinase inhibitors
CA2731498A1 (en) 2008-07-23 2010-01-28 Vertex Pharmaceuticals Incorporated Tri-cyclic pyrazolopyridine kinase inhibitors
CN102159546A (en) 2008-08-06 2011-08-17 沃泰克斯药物股份有限公司 Aminopyridine kinase inhibitors
US20110189685A1 (en) * 2008-10-22 2011-08-04 Cedars-Sinai Medical Center Methods of using jak3 genetic variants to diagnose and predict crohn's disease
US20110229471A1 (en) * 2008-11-26 2011-09-22 Cedars-Sinai Medical Center Methods of determining responsiveness to anti-tnf alpha therapy in inflammatory bowel disease
US9580752B2 (en) 2008-12-24 2017-02-28 Cedars-Sinai Medical Center Methods of predicting medically refractive ulcerative colitis (MR-UC) requiring colectomy
WO2010129668A1 (en) 2009-05-06 2010-11-11 Vertex Pharmaceuticals Incorporated Pyrazolopyridines
CA2768373A1 (en) * 2009-07-21 2011-01-27 The Board Of Trustees Of The Leland Stanford Junior University Heteroaryl benzamides, compositions and methods of use
WO2011017120A1 (en) * 2009-07-27 2011-02-10 Cedars-Sinai Medical Center Use of ccr9, ccl25, batf and il17/il23 pathway variants to diagnose and treat inflammatory bowel disease
JP2013518113A (en) 2010-01-27 2013-05-20 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazolopyrazine kinase inhibitor
EP2528917B1 (en) 2010-01-27 2016-10-19 Vertex Pharmaceuticals Incorporated Pyrazolopyridines useful for the treatment of autoimmune, inflammatory or (hyper)proliferative diseases
CN102869664A (en) 2010-01-27 2013-01-09 沃泰克斯药物股份有限公司 Pyrazolopyrimidine kinase inhibitors
CA2822044C (en) 2010-12-16 2021-12-21 Allergan, Inc. Novel 1,2- bis-sulfonamide derivatives as chemokine receptor modulators
KR101967936B1 (en) 2010-12-16 2019-04-10 알러간, 인코포레이티드 Sulfur derivatives as chemokine receptor modulators
KR101303991B1 (en) 2011-03-18 2013-09-04 성균관대학교산학협력단 Pharmaceutical composition for preventing or treating anti-inflammatory disease or infectious disease comprising m-3M3FBS as an effective component
UA116189C2 (en) 2011-05-02 2018-02-26 Мілленніум Фармасьютікалз, Інк. FORMULATION FOR ANTI-α4β7 ANTIBODY
TWI638661B (en) 2011-05-02 2018-10-21 千禧製藥公司 FORMULATION FOR ANTI-α4β7 ANTIBODY
SI2748146T1 (en) * 2011-07-22 2017-10-30 Chemocentryx, Inc. A crystalline form of the sodium salt of 4-tert-butyl-n-(4-chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl)-benzene sulfonamide
AU2012287233B2 (en) 2011-07-22 2017-07-20 Chemocentryx, Inc. Polymorphic forms of the sodium salt of 4-tert-butyl-N-[4-chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-benzenesulfonamide
AU2012287234B2 (en) * 2011-07-22 2017-02-02 Chemocentryx, Inc. Polymorphic forms of the sodium salt of 4-tert- butyl -N-[4-chloro-2-(1-oxy-pyridine-4-carbonyl)-phenyl]-benzene sulfonamide
BR112014021148B1 (en) 2012-02-29 2022-07-26 Chemocentryx, Inc PYRAZOL-1-YL BENZENE SULFONAMIDES AS CCR9 ANTAGONISTS, THEIR COMPOSITION AND METHOD OF MODULATE CCR FUNCTION(9) IN A CELL
WO2013130962A1 (en) 2012-03-01 2013-09-06 Allergan, Inc. Benzofuran- 2 - sulfonamide derivatives as chemokine receptor modulators
WO2014008417A1 (en) 2012-07-04 2014-01-09 Concert Pharmaceuticals, Inc. Deuterated vercirnon
WO2014088113A1 (en) 2012-12-04 2014-06-12 Millennium Pharmaceuticals, Inc. Prophylactic or therapeutic method for sjogren's syndrome using specific ccr9 receptor inhibitors/antagonists
EP2970239B1 (en) 2013-03-12 2018-10-10 Allergan, Inc. Sulfonamide derivatives as chemokine receptor modulators
BR112015024752A2 (en) 2013-03-27 2017-07-18 Cedars Sinai Medical Center mitigation and reversal of fibrosis and inflammation through inhibition of tl1a function and related signaling pathways
EP4105236A1 (en) 2013-07-19 2022-12-21 Cedars-Sinai Medical Center Anti-tl1a (tnfsf15) antibody for treatment of inflammatory bowel disease
AU2014372638A1 (en) * 2013-12-23 2016-06-16 Norgine B.V. Compounds useful as CCR9 modulators
JP2017503772A (en) 2013-12-23 2017-02-02 ノージン ビーブイ Benzenesulfonamide as a CCR9 inhibitor
MX2017004344A (en) 2014-10-06 2017-06-07 Chemocentryx Inc Combination therapy of inhibitors of c-c chemokine receptor type 9 (ccr9) and anti-alha4beta7 integrin blocking antibodies.
WO2016086147A1 (en) 2014-11-26 2016-06-02 Millennium Pharmaceuticals, Inc. Vedolizumab for the treatment of fistulizing crohn's disease
MA41636A (en) 2015-03-06 2018-01-09 Millennium Pharm Inc METHOD OF TREATMENT OF PRIMITIVE SCLEROSANT CHOLANGITIS
WO2017160700A1 (en) 2016-03-14 2017-09-21 Millennium Pharmaceuticals, Inc. Methods of treating or preventing graft versus host disease
EA201892071A1 (en) 2016-03-14 2019-03-29 Милленниум Фармасьютикалз, Инк. METHOD OF PREVENTION OF THE DISEASE "TRANSPLANTATE AGAINST THE OWNER"
US11186872B2 (en) 2016-03-17 2021-11-30 Cedars-Sinai Medical Center Methods of diagnosing inflammatory bowel disease through RNASET2
EP4134080A1 (en) 2016-11-23 2023-02-15 ChemoCentryx, Inc. Ccr2 inhibitors for use in treating renal diseases
US20200179486A1 (en) 2017-04-28 2020-06-11 Millennium Pharmaceuticals, Inc. Method of treating pediatric disorders
BR112020007183A2 (en) 2017-10-11 2020-09-24 Chemocentryx, Inc. treatment of segmented focal glomerulosclerosis with ccr2 antagonists
US10792360B1 (en) 2019-11-21 2020-10-06 Chemocentryx, Inc. Compositions and methods for treating inflammatory bowel disease using CCR9 inhibitor and anti-TNF-alpha blocking antibodies
US20210154292A1 (en) * 2019-11-21 2021-05-27 Chemocentryx, Inc. Compositions and Methods for Treating CCR9-Mediated Diseases using CCR9 Inhibitor and Anti-TNF-Alpha Blocking Antibodies

Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2997368A (en) * 1956-12-20 1961-08-22 Barent Mark Production of manganese hydroxide
US3121103A (en) * 1961-10-20 1964-02-11 Hoffmann La Roche Aminobenzophenones
US3344183A (en) * 1960-12-02 1967-09-26 Hoffmann La Roche 2 n-substituted amino benzophenones
US3442946A (en) * 1960-04-26 1969-05-06 Hoffmann La Roche 2-aminomercapto- and 2-aminoalkylcaptobenzophenone oximes
US3534062A (en) * 1967-05-16 1970-10-13 Upjohn Co Dibenzothiadiazocines
US3551427A (en) * 1969-05-09 1970-12-29 Sandoz Ag 4-thienyl-2-(1h)-quinazolones
US3842082A (en) * 1971-11-26 1974-10-15 Wander Ag Dr A 4-piperazinyl-10h-thieno(3,2-c)(1)benzazepines
US4166452A (en) * 1976-05-03 1979-09-04 Generales Constantine D J Jr Apparatus for testing human responses to stimuli
US4256108A (en) * 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4265874A (en) * 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US4372975A (en) * 1977-06-16 1983-02-08 Pierre Fabre Sa Substituted 2-benzoyl-4-chloroglycinanilide derivatives, a process for their production, and their use as medicaments
US4954518A (en) * 1987-10-08 1990-09-04 Toyama Chemical Company, Ltd. 4H-1-benzopyran-4-one derivative or its salt, process for producing the same and pharmaceutical composition comprising the same as active ingredient
US4992091A (en) * 1986-07-11 1991-02-12 The Dow Chemical Company Substituted pyrazolo[1,5-a]pyrimidine-2-sulfonamides and compositions and methods of controlling undesired vegetation
US4997940A (en) * 1986-07-11 1991-03-05 The Dow Chemical Company Pyrazolo[1,5-a]pyrimidine-2-sulfonyl chloride and -2-sulfide compounds
US5021591A (en) * 1986-07-11 1991-06-04 The Dow Chemical Company Pyrazolo[1,5-a]pyrimidine-2-sulfide compounds
US5071468A (en) * 1988-12-23 1991-12-10 Shell Internationale Research Maatschappij B.V. Triazolopyrimidine herbicides
US5093364A (en) * 1988-08-24 1992-03-03 Schering Agrochemicals Limited 5-fluoroanthranilic fungicides
US5155121A (en) * 1990-08-16 1992-10-13 Bayer Aktiengesellschaft Phenylsulphonamide substituted pyridinealkene- and aminooxyalkanecarboxylic acid derivatives
US5163995A (en) * 1988-05-25 1992-11-17 Dowelanco Herbicidal alkoxy-1,2,4-triazolo[1,5-c]pyrimidine-2-sulfonamides
US5185348A (en) * 1990-08-16 1993-02-09 Bayer Aktiengesellschaft Phenylsulphonamide substituted pyridinealkene- and -aminooxyalkanecarboxylic acid derivatives
US5217976A (en) * 1991-05-31 1993-06-08 Imperial Chemical Industries Plc 1,6-naphthyridone derivatives having angiotensin ii antagonist activity
US5217521A (en) * 1989-12-22 1993-06-08 Ciba-Geigy Corporation Triazolylsulfonamides
US5236937A (en) * 1991-02-11 1993-08-17 Imperial Chemical Industries Plc Pyridinyl compounds which are useful as angiotensin ii antagonists
US5292740A (en) * 1991-06-13 1994-03-08 Hoffmann-La Roche Inc. Sulfonamides
US5338755A (en) * 1990-07-31 1994-08-16 Elf Sanofi N-sulfonylindoline derivatives, their preparation and the pharmaceutical compositions in which they are present
US5420129A (en) * 1992-12-10 1995-05-30 Hoffmann-La Roche Inc. Phenylsulfonylamide pyrimidine
US5481005A (en) * 1990-07-31 1996-01-02 Sanofi N-sulfonylindoline derivatives, their preparation and the pharmaceutical compositions in which they are present
US5541186A (en) * 1993-06-28 1996-07-30 Hoffmann-La Roche Inc. Sulfonylaminopyrimidines
US5571775A (en) * 1994-07-11 1996-11-05 Dowelanco N-aryl[1,2,4]triazolo[1,5-a]pyridine-2-sulfonamide herbicides
US5589478A (en) * 1993-12-17 1996-12-31 Tanabe Seiyaku Co., Ltd. Benzenesulfonamide derivative and process for preparing thereof
US5739333A (en) * 1995-05-16 1998-04-14 Tanabe Seiyaku Co., Ltd. Sulfonamide derivative and process for preparing the same
US5780488A (en) * 1996-04-03 1998-07-14 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5883092A (en) * 1995-09-06 1999-03-16 Kowa Co., Ltd. Pyrimidine derivatives as endothelin antagonists
US5973148A (en) * 1996-11-13 1999-10-26 Dow Agrosciences Llc N-arylsulfilimine compounds and their use as catalysts in the preparation of N-arylarylsulfonamide compounds
US6136971A (en) * 1998-07-17 2000-10-24 Roche Colorado Corporation Preparation of sulfonamides
US6200995B1 (en) * 1998-01-29 2001-03-13 Tularik Inc. PPAR-γ modulators
US6297239B1 (en) * 1997-10-08 2001-10-02 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6297195B1 (en) * 1996-07-19 2001-10-02 Bayer Aktiengesellschaft Substituted triazoloazine sulphonamides
US6316450B1 (en) * 1997-07-11 2001-11-13 Smithkline Beecham P.L.C. Compounds
US20020009116A1 (en) * 2000-03-17 2002-01-24 Fujitsu Limited Complex coupling MQW semiconductor laser
US20020013314A1 (en) * 2000-02-01 2002-01-31 Bing-Yan Zhu 3,4-dihydro-2H-benzo[1,4]oxazine inhibitors of factor Xa
US20020012680A1 (en) * 1999-02-26 2002-01-31 Patel Mahesh V. Compositions and methods for improved delivery of lipid regulating agents
US20020037928A1 (en) * 2000-05-03 2002-03-28 Jaen Juan C. Combination therapeutic compositions and method of use
US20020037905A1 (en) * 1997-04-22 2002-03-28 Dahl Bjarne H. Substituted phenyl derivatives, their preparation and use
US20020052363A1 (en) * 2000-02-18 2002-05-02 Dinsmore Christopher J. Inhibitors of prenyl-protein transferase
US20020065303A1 (en) * 2000-02-01 2002-05-30 Bing-Yan Zhu Bivalent phenylene inhibitors of factor Xa
US6403607B1 (en) * 1998-03-30 2002-06-11 Hiroyoshi Hidaka Sulfonamide derivatives and drugs containing the same as the active ingredient
US20020072530A1 (en) * 2000-02-01 2002-06-13 Bing-Yan Zhu Indole and benzimidazole inhibitors of factor Xa
US20020103202A1 (en) * 2000-06-23 2002-08-01 Pinto Donald J.P. Heteroaryl-phenyl substituted factor Xa inhibitors
US6432624B1 (en) * 2000-05-08 2002-08-13 Fuji Photo Film Co., Ltd. Method of processing silver halide color photographic lightsensitive material
US20030060460A1 (en) * 1996-12-18 2003-03-27 Shuichi Ohuchida Sulfonamide and carboxamide derivatives and drugs containing the same as the active ingredient
US20030139390A1 (en) * 1999-06-30 2003-07-24 Tularik Inc. Compounds for the modulation of PPARgamma activity
US20040167113A1 (en) * 2002-11-18 2004-08-26 Solomon Ugashe Bis-aryl sulfonamides
US20050137193A1 (en) * 2002-11-18 2005-06-23 Chemocentreyx Aryl sulfonamides
US7238717B2 (en) * 2002-05-24 2007-07-03 Millennium Pharmaceuticals, Inc. CCR9 inhibitors and methods of use thereof

Family Cites Families (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB884847A (en) * 1958-04-14 1961-12-20 Ciba Ltd Mercapto compounds
DE1145626B (en) * 1959-12-10 1963-03-21 Hoffmann La Roche Process for the preparation of 3H-1, 4-Benzodiazepin-2 (1H) -one derivatives
CH418342A (en) * 1959-12-10 1966-08-15 Hoffmann La Roche Process for the preparation of 1,4-benzodiazepines
US3402171A (en) * 1960-12-02 1968-09-17 Hoffmann La Roche Process for preparing 5-aryl-3h-1, 4-benzodiazepin-2(1h)-ones
NL124621C (en) * 1962-11-13
CH561703A5 (en) 1970-12-11 1975-05-15 Hoffmann La Roche 7-Iodobenzodiazepin-2-ones - sedatives etc prepd by cyclising iodobenzophenones with glycine (esters)
GB1332697A (en) 1970-12-21 1973-10-03 Hoffmann La Roche Benzodiazepine derivatives
US3846477A (en) * 1972-09-20 1974-11-05 Robins Co Inc A H 2-alkylamino benzophenones
CH585222A5 (en) 1973-02-14 1977-02-28 Wander Ag Dr A Prepn. of antipsychotic heterocyclic piperazine derivs. - by reaction of heterocyclic ketones with metal piperazine complexes
FR2449677B1 (en) 1979-02-26 1986-03-28 Fabre Sa Pierre NOVEL SUBSTITUTED BENZOYL-2 GLYCYLANILIDES DERIVATIVES, THEIR PREPARATION AND THEIR APPLICATION AS ANXIOLYTIC DRUGS
US4312996A (en) * 1980-12-29 1982-01-26 Schering Corporation Benzodiazepine intermediates
DD251126A1 (en) 1984-12-21 1987-11-04 Univ Berlin Humboldt METHOD FOR PRODUCING NEW 2-ARYLSULFONAMIDO-BENZO AND -ACETOPHENONE AND THEIR OXIMES, PROCESS FOR THEIR PREPARATION AND THEIR USE IN MEDICAMENTS
JPH01156953A (en) * 1987-09-01 1989-06-20 Hokko Chem Ind Co Ltd Benzenesulfonanilide derivative and germicide for agriculture and horticulture
DE3825041A1 (en) 1988-07-20 1990-02-15 Schering Ag Pyrido[3,2-e][1,2,4]triazolo[1,5-a]pyrimidine-2-sulphonamides, processes for their preparation, and their use as herbicides, plant growth regulators and fungicides
FR2665441B1 (en) * 1990-07-31 1992-12-04 Sanofi Sa N-SULFONYL INDOLINE DERIVATIVES, THEIR PREPARATION, THE PHARMACEUTICAL COMPOSITIONS CONTAINING SAME.
FR2679903B1 (en) * 1991-08-02 1993-12-03 Elf Sanofi DERIVATIVES OF N-SULFONYL INDOLINE CARRYING AN AMIDIC FUNCTION, THEIR PREPARATION, THE PHARMACEUTICAL COMPOSITIONS CONTAINING SAME.
JPH04248984A (en) * 1991-02-05 1992-09-04 Kuraray Co Ltd Superoxide dismutase derivative and production thereof
JPH06145145A (en) 1991-12-27 1994-05-24 Ishihara Sangyo Kaisha Ltd Aminotrifluoromethylpyridine derivative or its salt, production thereof and phospholipase a2 inhibitor, antiinflammatory agent and antipancreatitic agent comprising the same
JPH06135934A (en) * 1991-12-27 1994-05-17 Ishihara Sangyo Kaisha Ltd Phospholipase a2 inhibitor, anti-inflammatory agent of anti-pancreatitic agent containing pyridine derivative or its salt
DK0556673T3 (en) 1992-02-15 1998-04-14 Hoechst Ag o-Substituted benzoylguanidines for the treatment and prophylaxis of cardiac arrhythmias and ischemic induced lesions and for cell proliferation inhibition
JP3229668B2 (en) 1992-09-25 2001-11-19 株式会社リコー Telelighting device
NZ250916A (en) 1993-02-27 1995-08-28 Nihon Nohyaku Co Ltd N-heteroaryl-n'-phenylureas, their use as acat inhibitors
US5514555A (en) 1993-03-12 1996-05-07 Center For Blood Research, Inc. Assays and therapeutic methods based on lymphocyte chemoattractants
WO1995033462A1 (en) 1994-06-02 1995-12-14 Smithkline Beecham Corporation Anti-inflammatory compounds
JPH089412A (en) 1994-06-22 1996-01-12 Nec Eng Ltd Automatic chrominance signal correcting circuit
FR2724654B1 (en) 1994-09-16 1997-12-12 Roussel Uclaf NEW GALLIC ACID DERIVATIVES, THEIR PREPARATION PROCESS, THE NEW INTERMEDIATES OBTAINED, THEIR APPLICATION AS MEDICAMENTS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
GB9602166D0 (en) 1996-02-02 1996-04-03 Zeneca Ltd Aminoheterocyclic derivatives
WO1998011218A1 (en) 1996-09-10 1998-03-19 Theodor-Kocher Institute Cxcr3 chemokine receptor, antibodies, nucleic acids, and methods of use
SE9604786D0 (en) 1996-12-20 1996-12-20 Astra Pharma Inc New compounds
AU7371998A (en) 1997-05-07 1998-11-27 University Of Pittsburgh Inhibitors of protein isoprenyl transferases
BR9814327A (en) 1997-12-23 2000-10-03 Warner Lambert Co Thiourea and benzamide compounds, compositions and processes for the treatment or prevention of inflammatory diseases and arteriosclerosis
GB9803411D0 (en) 1998-02-18 1998-04-15 Smithkline Beecham Plc Novel compounds
JP2002512957A (en) 1998-04-27 2002-05-08 スミスクライン・ビーチャム・コーポレイション CCR-3 receptor antagonist
AU3665199A (en) 1998-04-29 1999-11-16 Vertex Pharmaceuticals Incorporated Inhibitors of impdh enzyme
WO2000005214A2 (en) 1998-07-24 2000-02-03 Pfizer Inc. Isoquinolines as urokinase inhibitors
US6329159B1 (en) * 1999-03-11 2001-12-11 Millennium Pharmaceuticals, Inc. Anti-GPR-9-6 antibodies and methods of identifying agents which modulate GPR-9-6 function
KR100722344B1 (en) 1999-04-15 2007-05-29 브리스톨-마이어스스퀴브컴파니 Cyclic Protein Tyrosine Kinase Inhibitors
DK1192137T3 (en) 1999-06-30 2013-11-11 Amgen Inc Compounds for modulating PPAR gamma activity
WO2001009097A1 (en) 1999-07-30 2001-02-08 Vertex Pharmaceuticals Incorporated Cyclic amine derivatives for the treatment of neurological diseases
WO2001016114A2 (en) * 1999-08-27 2001-03-08 Chemocentryx, Inc. Heterocyclic compounds and methods for modulating cxcr3 function
AU6870500A (en) 1999-09-06 2001-04-10 Takeda Chemical Industries Ltd. Process for the preparation of 2,3-dihydroazepine derivatives
AU780787B2 (en) 1999-09-17 2005-04-14 Millennium Pharmaceuticals, Inc. Benzamides and related inhibitors of factor Xa
JP2003509412A (en) 1999-09-17 2003-03-11 シーオーアール セラピューティクス インコーポレイテッド Factor Xa inhibitor
JP2001089412A (en) 1999-09-22 2001-04-03 Otsuka Pharmaceut Co Ltd Benzene derivative or its pharmaceutically acceptable salt
GB9926302D0 (en) 1999-11-05 2000-01-12 Smithkline Beecham Plc Novel compounds
NZ520299A (en) 2000-02-03 2004-05-28 Eisai Co Ltd Integrin expression inhibitors containing sulphonamide derivatives
GB2359078A (en) * 2000-02-11 2001-08-15 Astrazeneca Uk Ltd Pharmaceutically active pyrimidine derivatives
NZ515442A (en) 2000-02-18 2004-03-26 Arch Dev Corp Polyhydroxylated benzene-containing compounds useful in reducing endocrine levels
CO5300399A1 (en) * 2000-02-25 2003-07-31 Astrazeneca Ab HETEROCICLIOCS CONTAINING NITROGEN, PROCESS FOR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
GB0011838D0 (en) * 2000-05-17 2000-07-05 Astrazeneca Ab Chemical compounds
HUP0301482A3 (en) 2000-06-28 2005-12-28 Japan Tobacco Inc Quinolinnyl and benzothiazolyl ppar-gamma modulators pharmaceutical compositions containing them and their use
EP1314721A1 (en) 2000-08-31 2003-05-28 Wakunaga Pharmaceutical Co., Ltd. Novel propenohydroxamic acid derivatives
CA2425259A1 (en) 2000-10-11 2002-04-18 Tularik, Inc. Modulation of ccr4 function
US20020147198A1 (en) 2001-01-12 2002-10-10 Guoqing Chen Substituted arylamine derivatives and methods of use
AR032230A1 (en) 2001-01-16 2003-10-29 Sumitomo Chem Takeda Agro Co SULFONAMIDE DERIVATIVE CONTAINING AN AGRICULTURAL AND HORTICALLY COMPOSITION
MXPA03006666A (en) 2001-01-25 2004-05-31 Guilford Pharm Inc Trisubstituted carbocyclic cyclophilin binding compounds and their use.
CN100406007C (en) 2002-12-20 2008-07-30 安姆根有限公司 Asthma and allergic inflammation modulators
US6993400B2 (en) * 2003-03-07 2006-01-31 Xerox Corporation System and method for real-time assignment of jobs to production cells
WO2004099127A1 (en) 2003-05-07 2004-11-18 Novo Nordisk A/S Novel compounds as kinase inhibitors
US7393873B2 (en) 2003-07-02 2008-07-01 Merck & Co., Inc. Arylsulfonamide derivatives
PL2354126T3 (en) 2005-01-14 2014-03-31 Chemocentryx Inc Heteroaryl sulfonamides and CCR2
JP4364168B2 (en) 2005-07-11 2009-11-11 アルゼ株式会社 Game machine
WO2007014054A2 (en) 2005-07-22 2007-02-01 Glaxo Group Limted Benzenesulfonamide inhibitor of ccr2 chemokine receptor

Patent Citations (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2997368A (en) * 1956-12-20 1961-08-22 Barent Mark Production of manganese hydroxide
US3442946A (en) * 1960-04-26 1969-05-06 Hoffmann La Roche 2-aminomercapto- and 2-aminoalkylcaptobenzophenone oximes
US3344183A (en) * 1960-12-02 1967-09-26 Hoffmann La Roche 2 n-substituted amino benzophenones
US3121103A (en) * 1961-10-20 1964-02-11 Hoffmann La Roche Aminobenzophenones
US3534062A (en) * 1967-05-16 1970-10-13 Upjohn Co Dibenzothiadiazocines
US3551427A (en) * 1969-05-09 1970-12-29 Sandoz Ag 4-thienyl-2-(1h)-quinazolones
US3842082A (en) * 1971-11-26 1974-10-15 Wander Ag Dr A 4-piperazinyl-10h-thieno(3,2-c)(1)benzazepines
US4166452A (en) * 1976-05-03 1979-09-04 Generales Constantine D J Jr Apparatus for testing human responses to stimuli
US4256108A (en) * 1977-04-07 1981-03-17 Alza Corporation Microporous-semipermeable laminated osmotic system
US4372975A (en) * 1977-06-16 1983-02-08 Pierre Fabre Sa Substituted 2-benzoyl-4-chloroglycinanilide derivatives, a process for their production, and their use as medicaments
US4265874A (en) * 1980-04-25 1981-05-05 Alza Corporation Method of delivering drug with aid of effervescent activity generated in environment of use
US5021591A (en) * 1986-07-11 1991-06-04 The Dow Chemical Company Pyrazolo[1,5-a]pyrimidine-2-sulfide compounds
US4997940A (en) * 1986-07-11 1991-03-05 The Dow Chemical Company Pyrazolo[1,5-a]pyrimidine-2-sulfonyl chloride and -2-sulfide compounds
US4992091A (en) * 1986-07-11 1991-02-12 The Dow Chemical Company Substituted pyrazolo[1,5-a]pyrimidine-2-sulfonamides and compositions and methods of controlling undesired vegetation
US4954518A (en) * 1987-10-08 1990-09-04 Toyama Chemical Company, Ltd. 4H-1-benzopyran-4-one derivative or its salt, process for producing the same and pharmaceutical composition comprising the same as active ingredient
US5163995A (en) * 1988-05-25 1992-11-17 Dowelanco Herbicidal alkoxy-1,2,4-triazolo[1,5-c]pyrimidine-2-sulfonamides
US5093364A (en) * 1988-08-24 1992-03-03 Schering Agrochemicals Limited 5-fluoroanthranilic fungicides
US5071468A (en) * 1988-12-23 1991-12-10 Shell Internationale Research Maatschappij B.V. Triazolopyrimidine herbicides
US5217521A (en) * 1989-12-22 1993-06-08 Ciba-Geigy Corporation Triazolylsulfonamides
US5338755A (en) * 1990-07-31 1994-08-16 Elf Sanofi N-sulfonylindoline derivatives, their preparation and the pharmaceutical compositions in which they are present
US5481005A (en) * 1990-07-31 1996-01-02 Sanofi N-sulfonylindoline derivatives, their preparation and the pharmaceutical compositions in which they are present
US5155121A (en) * 1990-08-16 1992-10-13 Bayer Aktiengesellschaft Phenylsulphonamide substituted pyridinealkene- and aminooxyalkanecarboxylic acid derivatives
US5185348A (en) * 1990-08-16 1993-02-09 Bayer Aktiengesellschaft Phenylsulphonamide substituted pyridinealkene- and -aminooxyalkanecarboxylic acid derivatives
US5236937A (en) * 1991-02-11 1993-08-17 Imperial Chemical Industries Plc Pyridinyl compounds which are useful as angiotensin ii antagonists
US5217976A (en) * 1991-05-31 1993-06-08 Imperial Chemical Industries Plc 1,6-naphthyridone derivatives having angiotensin ii antagonist activity
US5294620A (en) * 1991-05-31 1994-03-15 Imperial Chemical Industries Plc 1,6-naphthyridinone derivatives having angiotension II antagonist activity
US5292740A (en) * 1991-06-13 1994-03-08 Hoffmann-La Roche Inc. Sulfonamides
US5420129A (en) * 1992-12-10 1995-05-30 Hoffmann-La Roche Inc. Phenylsulfonylamide pyrimidine
US5541186A (en) * 1993-06-28 1996-07-30 Hoffmann-La Roche Inc. Sulfonylaminopyrimidines
US5589478A (en) * 1993-12-17 1996-12-31 Tanabe Seiyaku Co., Ltd. Benzenesulfonamide derivative and process for preparing thereof
US5571775A (en) * 1994-07-11 1996-11-05 Dowelanco N-aryl[1,2,4]triazolo[1,5-a]pyridine-2-sulfonamide herbicides
US5739333A (en) * 1995-05-16 1998-04-14 Tanabe Seiyaku Co., Ltd. Sulfonamide derivative and process for preparing the same
US5883092A (en) * 1995-09-06 1999-03-16 Kowa Co., Ltd. Pyrimidine derivatives as endothelin antagonists
US5780488A (en) * 1996-04-03 1998-07-14 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6297195B1 (en) * 1996-07-19 2001-10-02 Bayer Aktiengesellschaft Substituted triazoloazine sulphonamides
US5973148A (en) * 1996-11-13 1999-10-26 Dow Agrosciences Llc N-arylsulfilimine compounds and their use as catalysts in the preparation of N-arylarylsulfonamide compounds
US20030060460A1 (en) * 1996-12-18 2003-03-27 Shuichi Ohuchida Sulfonamide and carboxamide derivatives and drugs containing the same as the active ingredient
US20020037905A1 (en) * 1997-04-22 2002-03-28 Dahl Bjarne H. Substituted phenyl derivatives, their preparation and use
US6316450B1 (en) * 1997-07-11 2001-11-13 Smithkline Beecham P.L.C. Compounds
US6297239B1 (en) * 1997-10-08 2001-10-02 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6200995B1 (en) * 1998-01-29 2001-03-13 Tularik Inc. PPAR-γ modulators
US6403607B1 (en) * 1998-03-30 2002-06-11 Hiroyoshi Hidaka Sulfonamide derivatives and drugs containing the same as the active ingredient
US6136971A (en) * 1998-07-17 2000-10-24 Roche Colorado Corporation Preparation of sulfonamides
US20020012680A1 (en) * 1999-02-26 2002-01-31 Patel Mahesh V. Compositions and methods for improved delivery of lipid regulating agents
US20030139390A1 (en) * 1999-06-30 2003-07-24 Tularik Inc. Compounds for the modulation of PPARgamma activity
US6770648B2 (en) * 1999-06-30 2004-08-03 Tularik Inc. Compounds for the modulation of PPARγ activity
US20020065303A1 (en) * 2000-02-01 2002-05-30 Bing-Yan Zhu Bivalent phenylene inhibitors of factor Xa
US20020072530A1 (en) * 2000-02-01 2002-06-13 Bing-Yan Zhu Indole and benzimidazole inhibitors of factor Xa
US20020013314A1 (en) * 2000-02-01 2002-01-31 Bing-Yan Zhu 3,4-dihydro-2H-benzo[1,4]oxazine inhibitors of factor Xa
US20020052363A1 (en) * 2000-02-18 2002-05-02 Dinsmore Christopher J. Inhibitors of prenyl-protein transferase
US20020009116A1 (en) * 2000-03-17 2002-01-24 Fujitsu Limited Complex coupling MQW semiconductor laser
US20020037928A1 (en) * 2000-05-03 2002-03-28 Jaen Juan C. Combination therapeutic compositions and method of use
US6653332B2 (en) * 2000-05-03 2003-11-25 Tularik Inc. Combination therapeutic compositions and method of use
US6432624B1 (en) * 2000-05-08 2002-08-13 Fuji Photo Film Co., Ltd. Method of processing silver halide color photographic lightsensitive material
US20020103202A1 (en) * 2000-06-23 2002-08-01 Pinto Donald J.P. Heteroaryl-phenyl substituted factor Xa inhibitors
US7238717B2 (en) * 2002-05-24 2007-07-03 Millennium Pharmaceuticals, Inc. CCR9 inhibitors and methods of use thereof
US7282502B2 (en) * 2002-05-24 2007-10-16 Millennium Pharmaceuticals, Inc. CCR9 inhibitors and methods of use thereof
US20040167113A1 (en) * 2002-11-18 2004-08-26 Solomon Ugashe Bis-aryl sulfonamides
US20040171654A1 (en) * 2002-11-18 2004-09-02 Solomon Ugashe Aryl sulfonamides
US20050137193A1 (en) * 2002-11-18 2005-06-23 Chemocentreyx Aryl sulfonamides
US6939885B2 (en) * 2002-11-18 2005-09-06 Chemocentryx Aryl sulfonamides
US7582661B2 (en) * 2002-11-18 2009-09-01 Chemocentryx, Inc. Aryl sulfonamides

Also Published As

Publication number Publication date
US20070066823A1 (en) 2007-03-22
JP4828822B2 (en) 2011-11-30
US9035096B2 (en) 2015-05-19
JP2014132018A (en) 2014-07-17
ZA200409131B (en) 2005-09-28
EP1507756B1 (en) 2015-07-22
US7282502B2 (en) 2007-10-16
US20090054495A1 (en) 2009-02-26
JP2011105725A (en) 2011-06-02
WO2003099773A1 (en) 2003-12-04
US7238717B2 (en) 2007-07-03
NZ536504A (en) 2008-04-30
JP2010047584A (en) 2010-03-04
JP5432221B2 (en) 2014-03-05
EP1507756A4 (en) 2006-03-15
CA2485681C (en) 2012-10-16
JP5548169B2 (en) 2014-07-16
US20060167251A1 (en) 2006-07-27
JP4818798B2 (en) 2011-11-16
JP2011213740A (en) 2011-10-27
AU2003248549B2 (en) 2010-04-08
US8569543B2 (en) 2013-10-29
US20140107160A1 (en) 2014-04-17
JP2006265259A (en) 2006-10-05
AU2003248549A1 (en) 2003-12-12
US7999109B2 (en) 2011-08-16
EP1507756A1 (en) 2005-02-23
AU2010201073B2 (en) 2011-09-29
US20110313000A1 (en) 2011-12-22
EP2402309A1 (en) 2012-01-04
JP2005526857A (en) 2005-09-08
US20100093802A1 (en) 2010-04-15
EP2399903A1 (en) 2011-12-28
US20160068479A1 (en) 2016-03-10
EP2402310A1 (en) 2012-01-04
CA2784937A1 (en) 2003-12-04
JP2011246486A (en) 2011-12-08
MXPA04011465A (en) 2005-02-14
US7659272B2 (en) 2010-02-09
US7820717B2 (en) 2010-10-26
US20040038976A1 (en) 2004-02-26
US8030517B2 (en) 2011-10-04
NZ566263A (en) 2009-09-25
JP5544283B2 (en) 2014-07-09
US20080103180A1 (en) 2008-05-01
AU2010201073A1 (en) 2010-04-08
CA2485681A1 (en) 2003-12-04
US20100280074A1 (en) 2010-11-04

Similar Documents

Publication Publication Date Title
US7999109B2 (en) CCR9 inhibitors and methods of use thereof
AU2011218661B2 (en) CCR9 inhibitors and methods of use thereof
AU2007202607B2 (en) Substituted Pyridinones as Modulators of p38 MAP Kinase

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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