WO2004103279A2 - 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists - Google Patents

3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists Download PDF

Info

Publication number
WO2004103279A2
WO2004103279A2 PCT/US2004/014837 US2004014837W WO2004103279A2 WO 2004103279 A2 WO2004103279 A2 WO 2004103279A2 US 2004014837 W US2004014837 W US 2004014837W WO 2004103279 A2 WO2004103279 A2 WO 2004103279A2
Authority
WO
WIPO (PCT)
Prior art keywords
disease
group
halo
mmol
compound according
Prior art date
Application number
PCT/US2004/014837
Other languages
French (fr)
Other versions
WO2004103279A3 (en
Inventor
Vincent J. Colandrea
George A. Doherty
Jeffrey J. Hale
Christopher Lynch
Sander G. Mills
William Edward Neway, Iii
Leslie Toth
Original Assignee
Merck & Co., 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 Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to JP2006532984A priority Critical patent/JP2006528980A/en
Priority to EP04751981A priority patent/EP1625123A4/en
Priority to CA002524867A priority patent/CA2524867A1/en
Priority to AU2004240586A priority patent/AU2004240586A1/en
Priority to US10/554,665 priority patent/US20060252741A1/en
Publication of WO2004103279A2 publication Critical patent/WO2004103279A2/en
Publication of WO2004103279A3 publication Critical patent/WO2004103279A3/en

Links

Classifications

    • 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
    • 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
    • A61P1/18Drugs 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
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • 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
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • 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
    • A61P3/06Antihyperlipidemics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • 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

Definitions

  • the present invention is related to compounds that are SlPi/Edgl receptor agonists and thus have immunosuppressive activities by modulating leukocyte trafficking, sequestering lymphocytes in secondary lymphoid tissues, and interfering with celkcell interactions required for an efficient immune response.
  • the invention is also directed to pharmaceutical compositions containing such compounds and methods of treatment or prevention.
  • Immunosuppressive agents have been shown to be useful in a wide variety of autoimmune and chronic inflammatory diseases, including systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis and other disorders such as Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy, atopic dermatitis and asthma.
  • chemotherapeutic regimens for the treatment of cancers, lymphomas and leukemias.
  • pathogenesis of each of these conditions may be quite different, they have in common the appearance of a variety of autoantibodies and/or self-reactive lymphocytes. Such self-reactivity may be due, in part, to a loss of the homeostatic controls under which the normal immune system operates.
  • the host lymphocytes recognize the foreign tissue antigens and begin to produce both cellular and humoral responses including antibodies, cytokines and cytotoxic lymphocytes which lead to graft rejection.
  • autoimmune or a rejection process tissue destruction caused by inflammatory cells and the mediators they release.
  • Anti- inflammatory agents such as NSAIDs act principally by blocking the effect or secretion of these mediators but do nothing to modify the immunologic basis of the disease.
  • cytotoxic agents such as cyclophosphamide, act in such a nonspecific fashion that both the normal and autoimmune responses are shut off. Indeed, patients treated with such nonspecific immunosuppressive agents are as likely to succumb to infection as they are to their autoimmune disease.
  • Cyclosporin A is a drag used to prevent rejection of transplanted organs.
  • FK-506 is another drug approved for the prevention of transplant organ rejection, and in particular, liver transplantation. Cyclosporin A and FK-506 act by inhibiting the body's immune system from mobilizing its vast arsenal of natural protecting agents to reject the transplant's foreign protein. Cyclosporin A was approved for the treatment of severe psoriasis and has been approved by European regulatory agencies for the treatment of atopic dermatitis. Though they are effective in delaying or suppressing transplant rejection, Cyclosporin A and FK-506 are known to cause several undesirable side effects including nephrotoxicity, neurotoxicity, and gastrointestinal discomfort. Therefore, an immunosuppressant without these side effects still remains to be developed and would be highly desirable.
  • the immunosuppressive compound FTY720 is a lymphocyte sequestration agent cunently in clinical trials.
  • FTY720 is metabolized in mammals to a compound that is a potent agonist of sphingosine 1 -phosphate receptors.
  • Agonism of sphingosine 1 -phosphate receptors modulates leukocyte trafficking, induces the sequestration of lymphocytes (T-cells and B-cells) in lymph nodes and Peyer's patches without lymphodepletion, and disrapts splenic architecture, thereby interfering with T cell dependent and independent antibody responses.
  • Such immunosuppression is desirable to prevent rejection after organ transplantation and in the treatment of autoimmune disorders.
  • Sphingosine 1 -phosphate is a bioactive sphingolipid metabolite that is secreted by hematopoietic cells and stored and released from activated platelets.
  • SIPi S1P2, SIP3, SIP4, and SIP5, also known as endothelial differentiation genes Edgl, Edg5, Edg3, Edg6, Edg8), that have widespread cellular and tissue distribution and are well conserved in human and rodent species (see Table). Binding to SIP receptors elicits signal transduction through Gq-, Gi/o, G12-, G13-, and Rho-dependent pathways. Ligand-induced activation of SIP 1 and SIP3 has been shown to promote angiogenesis, chemotaxis, and adherens junction assembly through Rac- and Rho-, see Lee, M.-J., S. Thangada, K.P.
  • SIP5 is primarily a neuronal receptor with some expression in lymphoid tissue, see hn, D.S., C.E. Heise, N. Ancellin, B.F. O'Dowd, G.J. Shei, R.P. Heavens, M.R. Rigby, T. Hla, S. Mandala, G. McAllister, S. George, and K.R. Lynch. 2000. J. Biol. C em. 275:14281-6.
  • Administration of sphingosine 1 -phosphate to animals induces systemic sequestration of peripheral blood lymphocytes into secondary lymphoid organs, thus resulting in therapeutically useful immunosuppression, see Mandala, S., R.
  • sphingosine 1-phosphate also has cardiovascular and bronchoconstrictor effects that limit its utility as a therapeutic agent. Intravenous administration of sphingosine 1-phosphate decreases the heart rate, ventricular contraction and blood pressure in rats, see Sugiyama, A., N.N. Aye, Y. Yatomi, Y.
  • sphingosine 1-phosphate modulates contraction, cell growth and cytokine production that promote bronchoconstriction, airway inflammation and remodeling in asthma, see Ammit, A.J., A.T. Hastie, L. C. Edsall, R.K. Hoffman, Y. Amrani, V.P. Krymskaya, S.A. Kane, S.P. Peters, R.B. Perm, S. Spiegel, RA. Panettieri. Jr. 2001, FASEB J. 15:1212-1214.
  • the undesirable effects of sphingosine 1-phosphate are associated with its non-selective, potent agonist activity on all SIP receptors.
  • the present invention encompasses compounds which are agonists of the SlPi/Edgl receptor having selectivity over the SlP3/Edg3 receptor.
  • An SlPl/Edgl receptor selective agonist has advantages over cu ⁇ ent therapies and extends the therapeutic window of lymphocyte sequestration agents, allowing better tolerability with higher dosing and thus improving efficacy as monotherapy.
  • immunosuppressants While the main use for immunosuppressants is in treating bone ma ⁇ ow, organ and transplant rejection, other uses for such compounds include the treatment of arthritis, in particular, rheumatoid arthritis, insulin and non-insulin dependent diabetes, multiple sclerosis, psoriasis, inflammatory bowel disease, Crohn's disease, lupus erythematosis and the like.
  • the present invention is focused on providing immunosuppressant compounds that are safer and more effective than prior compounds.
  • the present invention encompasses compounds of Formula I:
  • the compounds are useful for treating immune mediated diseases and conditions, such as bone manow, organ and tissue transplant rejection.
  • Pharmaceutical compositions and methods of use are included.
  • A is C-R3 orN
  • D is C-R orN
  • E is C-R6 orN and
  • G is C-R7 orN
  • X, Y and Z are independently selected from the group consisting of: N and C-R8, with the proviso that at least one of X, Y and Z is not N;
  • Rl and R2 are each independently selected from the group consisting of:
  • Ci-6alkyl optionally substituted with 1 to 3 halo groups, or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form a 3- to 6-membered saturated monocyclic ring;
  • R3, R4, R6 and R7 are each independently selected from the group consisting of:
  • Ci-4alkyl or Ci-4alkoxy each optionally substituted with 1 to
  • R5 is selected from the group consisting of: (1) Ci-6alkyl,
  • Ci-4alkyl or Ci-4alkoxy each optionally substituted with oxo, hydroxy or 1 to 3 halo groups,
  • R4 and R5 may be joined together with the atoms to which they are attached to form a 5 or 6-membered monocyclic ring, optionally containing 1 to 3 heteratoms selected from O, S and NR8, said ring optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, Ci-4alkyl and Ci-4alkoxy, said Ci-4alkyl or Ci-4alkoxy optionally substituted with 1 to 3 halo groups;
  • each R8 is independently selected from the group consisting of: hydrogen, halo and Cl-4alkyl, wherein said Cl-4alkyl is optionally substituted with 1 to 3 halo groups;
  • HET is selected from the group consisting of: benzimidazolyl, benzofuranyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, py ⁇ olyl, quinazolinyl, quinolyl, quinoxalinyl, thiadiazolyl, thiazoly
  • A is N
  • E is C-R6 and
  • G is C-R7.
  • A is C-R3
  • E is C-R6 and
  • G is C-R7.
  • Another embodiment of the invention encompasses a compound of Formula I wherein:
  • A is C-R3
  • E is C-R6 and
  • G is C-R7;
  • X, Y and Z are C-R8.
  • A is C-R3
  • E is C-R6 and
  • G is C-R7;
  • R3, R6 and R7 are hydrogen. Within this embodiment is encompassed a compound of Formula I wherein R is trifluoromethyl or cyano.
  • A is C-R3
  • D is C-R4
  • E is C-R6
  • G is C-R7;
  • Rl and R2 are each independently selected from the group consisting of hydrogen, methyl and ethyl.
  • A is C-R3
  • E is C-R6 and
  • G is C-R7;
  • R5 is selected from the group consisting of:
  • A is C-R3
  • D is C-R4
  • E is C-R6 and
  • G is C-R7;
  • R5 is selected from the group consisting of:
  • phenyl optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, methyl, methoxy and hydroxymethyl,
  • A is C-R3
  • E is C-R6 and
  • G is C-R7;
  • X is N and Y and Z are both C-R8.
  • A is C-R3
  • D is C-R4
  • E is C-R6 and
  • G is C-R7;
  • A is C-R3
  • E is C-R6 and
  • G is C-R7;
  • Rl and R2 are each independently selected from the group consisting of: hydrogen and methyl
  • R3, R6 and R are hydrogen
  • R4 is trifluoromethyl or cyano
  • R5 is C2-6alkoxy, optionally substituted with 1 to 5 fluoro groups.
  • R5 is selected from 2,2,2- trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy.
  • R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy, X, Y and Z are C-R ⁇ and each R8 is independently selected from hydrogen, methyl and halo.
  • R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy
  • X is N and Y and Z are both C-R8 and each R8 is independently selected from hydrogen, methyl and halo.
  • R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy
  • X and Z are both C-R8 and Y is N and each Ro" is independently selected from hydrogen, methyl and halo.
  • the invention also encompasses a method of treating an immunoregulatory abnormality in a mammalian patient in need of such treatment comprising administering to said patient a compound of Formula I in an amount that is effective for treating said immunoregulatory abnormality.
  • the immunoregulatory abnormality is an autoimmune or chronic inflammatory disease selected from the group consisting of: systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy and asthma.
  • an autoimmune or chronic inflammatory disease selected from the group consisting of: systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid
  • the immunoregulatory abnormality is bone manow or organ transplant rejection or graft-versus-host disease.
  • the immunoregulatory abnormality is selected from the group consisting of: transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, post-infectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous de ⁇ natitis, sebonhoeic de ⁇ natitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa,
  • autoimmune syndromes including rheumato
  • the invention also encompasses a method of suppressing the immune system in a mammalian patient in need of immunosuppression comprising administering to said patient an immunosuppressmg effective amount of a compound of Formula I.
  • the invention also encompasses a pha ⁇ naceutical composition comprised of a compound of Formula I in combination with a pharmaceutically acceptable carrier.
  • the invention also encompasses a method of treating a respiratory disease or condition in a mammalian patient in need of such treatment comprising administering to said patient a compound of Formula I in an amount that is effective for treating said respiratory disease or condition.
  • the respiratory disease or condition is selected from the group consisting of: asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia.
  • halogen or halo includes F, CI, Br, and I.
  • alkyl means linear or branched structures and combinations thereof, having the indicated number of carbon atoms.
  • Ci-6alkyl includes methyl, ethyl, propyl, 2-propyl, s- and t-butyl, butyl, pentyl, hexyl, 1,1- dimethylethyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • alkoxy means alkoxy groups of a straight, branched or cyclic configuration having the indicated number of carbon atoms. Ci-6alkoxy, for example, includes methoxy, ethoxy, propoxy, isopropoxy, and the like.
  • alkylthio means alkylthio groups having the indicated number of carbon atoms of a straight, branched or cyclic configuration.
  • Ci- 6 a lkylthio for example, includes methylthio, propylthio, isopropylthio, and the like.
  • alkenyl means linear or branched structures and combinations thereof, of the indicated number of carbon atoms, having at least one carbon-to-carbon double bond, wherein hydrogen may be replaced by an additional carbon-to-carbon double bond.
  • C2-6alkenyl for example, includes ethenyl, propenyl, 1-methylethenyl, butenyl and the like.
  • alkynyl means linear or branched structures and combinations thereof, of the indicated number of carbon atoms, having at least one carbon-to-carbon triple bond.
  • C3_6alkynyl for example, includes , propenyl, 1- methylethenyl, butenyl and the like.
  • cycloalkyl means mono-, bi- or tri-cyclic structures, optionally combined with linear or branched structures, having the indicated number of carbon atoms.
  • cycloalkyl groups include cyclopropyl, cyclopentyl, cycloheptyl, adamantyl, cyclododecylmethyl, 2-ethyl-l- bicyclo[4.4.0]decyl, cyclobutylmethyl and the like.
  • cycloalkoxy means cycloalkyl-O- wherein cycloalkyl is as defined above.
  • cycloalkoxy includes cyclobutoxy.
  • acyl means an alkyl group as defined above substituted at the 1 -position with oxo. Examples include formyl, acetyl, propionyl, butyryl, valeryl and hexanoyl.
  • aryl is defined as a mono- or bi-cyclic aromatic ring system and includes, for example, phenyl, naphthyl, and the like.
  • aralkyl means an alkyl group as defined above of 1 to 6 carbon atoms with an aryl group as defined above substituted for one of the alkyl hydrogen atoms, for example, benzyl and the like.
  • aryloxy means an aryl group as defined above attached to a molecule by an oxygen atom (aryl-O) and includes, for example, phenoxy, naphthoxy and the like.
  • aralkoxy means an aralkyl group as defined above attached to a molecule by an oxygen atom (aralkyl-O) and includes, for example, benzyloxy, and the like.
  • arylthio is defined as an aryl group as defined above attached to a molecule by an sulfur atom (aryl-S) and includes, for example, thiophenyoxy, thionaphthoxy and the like.
  • aroyl means an aryl group as defined above attached to a molecule by an carbonyl group (aryl-C(O)-) and includes, for example, benzoyl, naphthoyl and the like.
  • aroyloxy means an aroyl group as defined above attached to a molecule by an oxygen atom (aroyl-O) and includes, for example, benzoyloxy or benzoxy, naphthoyloxy and the like.
  • treating encompasses not only treating a patient to relieve the patient of the signs and symptoms of the disease or condition but also prophylactically treating an asymptomatic patient to prevent the onset or progression of the disease or condition.
  • amount effective for treating is intended to mean that amount of a drag or pharmaceutical agent that will elicit the biological or medical response of a tissue, a system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.
  • amount of a pharmaceutical dmg that will prevent or reduce the risk of occunence of the biological or medical event that is sought to be prevented in a tissue, a system, animal or human by a researcher, veterinarian, medical doctor or other clinician.
  • compositions described herein includes pharmaceutically acceptable salts and hydrates.
  • Pharmaceutically acceptable salts include both the metallic (inorganic) salts and organic salts; a list of which is given in Remington's Pharmaceutical Sciences, 17th Edition, pg. 1418 (1985). It is well known to one skilled in the art that an appropriate salt form is chosen based on physical and chemical stability, flowability, hydroscopicity and solubility.
  • pharmaceutically acceptable salts include, but are not limited to salts of inorganic acids such as hydrochloride, sulfate, phosphate, diphosphate, hydrobromide, and nitrate or salts of an organic acid such as malate, maleate, fumarate, tartiate, succinate, citrate, acetate, lactate, methanesulfonate, p- toluenesulfonate or pamoate, salicylate and stearate.
  • pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium and ammonium (especially ammonium salts with secondary amines).
  • Prefe ⁇ ed salts of this invention for the reasons cited above include potassium, sodium, calcium and ammonium salts.
  • crystal forms, hydrates and solvates of the compounds of Formula I are crystal forms, hydrates and solvates of the compounds of Formula I.
  • pharmaceutically acceptable hydrate means the compounds of the instant invention crystallized with one or more molecules of water to form a hydrated form.
  • the invention also includes the compounds falling within Formula I in the form of one or more stereoisomers, in substantially pure form or in the form of a mixture of stereoisomers. All such isomers are encompassed within the present invention.
  • the compounds of the present invention are immunoregulatory agents useful for treating or preventing automimmune or chronic inflammatory diseases.
  • the compounds of the present invention are useful to suppress the immune system in instances where immunosuppression is in order, such as in bone manow, organ or transplant rejection, autoimmune and chronic inflammatory diseases, including systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy and asthma.
  • autoimmune and chronic inflammatory diseases including systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease,
  • the compounds of the present invention are useful to treat or prevent a disease or disorder selected from the group consisting of: transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomeralonephritis, post-infectious autoimmune diseases including rheumatic fever and post-infectious glomeralonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, sebo ⁇ hoeic de ⁇ natitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa,
  • the compounds of the present invention are also useful for treating or preventing Alzheimer's Disease.
  • Also embodied within the present invention is a method of preventing or treating resistance to transplantation or transplantation rejection of organs or tissues in a mammalian patient in need thereof, which comprises administering a therapeutically effective amount of the compound of Formula I.
  • a method of suppressing the immune system in a mammalian patient in need thereof, which comprises administering to the patient an immune system suppressing amount of the compound of Formula I is yet another embodiment.
  • the method described herein encompasses a method of treating or preventing bone manow or organ transplant rejection which is comprised of admininstering to a mammalian patient in need of such treatment or prevention a compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof, in an amount that is effective for treating or preventing bone manow or organ transplant rejection.
  • the compounds of the present invention are also useful for treating a respiratory dieases or condition, such as asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia
  • a respiratory dieases or condition such as asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia
  • the compounds of the present invention are selective agonists of the SlPi/Edgl receptor having selectivity over SlP3/Edg3 receptor.
  • An Edgl selective agonist has advantages over cu ⁇ ent therapies and extends the therapeutic window of lymphocytes sequestration agents, allowing better tolerability with higher dosing and thus improving efficacy as monotherapy.
  • the present invention also includes a pharmaceutical formulation comprising a pharmaceutically acceptable carrier and the compound of Formula I or a pharmaceutically acceptable salt or hydrate thereof.
  • a prefe ⁇ ed embodiment of the formulation is one where a second immunosuppressive agent is also included.
  • second immunosuppressive agents are, but are not limited to azathioprine, brequinar sodium, deoxyspergualin, mizaribine, mycophenolic acid mo ⁇ holino ester, cyclosporin, FK-506, rapamycin, FTY720 and ISAtx247 (Isotechnika).
  • the present compounds are useful in the treatment of autoimmune diseases, including the prevention of rejection of bone manow transplant, foreign organ transplants and/or related afflictions, diseases and illnesses.
  • the compounds of this invention can be administered by any means that effects contact of the active ingredient compound with the site of action in the body of a warm-blooded animal.
  • administration can be oral, topical, including tiansdermal, ocular, buccal, intianasal, inhalation, intravaginal, rectal, intracisternal and parenteral.
  • parenteral refers to modes of administration which include subcutaneous, intravenous, intramuscular, intraarticular injection or infusion, intrasternal and intraperitoneal.
  • the compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but are generally administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
  • the dosage administered will be dependent on the age, health and weight of the recipient, the extent of disease, kind of concu ⁇ ent treatment, if any, frequency of tieatment and the nature of the effect desired.
  • a daily dosage of active ingredient compound will be from about 0.1 -2000 milligrams per day.
  • the active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, troches, dragees, granules and powders, or in liquid dosage forms, such as elixirs, syrups, emulsions, dispersions, and suspensions.
  • the active ingredient can also be administered parenterally, in sterile liquid dosage forms, such as dispersions, suspensions or solutions.
  • dosages forms that can also be used to administer the active ingredient as an ointment, cream, drops, tiansdermal patch or powder for topical administration, as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration, as an aerosol spray or powder composition for inhalation or intranasal administration, or as a cream, ointment, spray or suppository for rectal or vaginal administration.
  • Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
  • water a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene gycols are suitable carriers for parenteral solutions.
  • Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances.
  • Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents.
  • citric acid and its salts and sodium EDTA are also used.
  • parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propylparaben, and chlorobutanol.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.
  • the compounds of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers.
  • the compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device.
  • the prefened delivery system for inhalation is a metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons.
  • MDI metered dose inhalation
  • an ophthalmic preparation may be formulated with an appropriate weight percent solution or suspension of the compounds of Formula I in an appropriate ophthalmic vehicle, such that the compound is maintained in contact with the ocular surface for a sufficient time period to allow the compound to penetrate the comeal and internal regions of the eye.
  • a large number of unit capsules are prepared by filling standard two- piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
  • SOFT GELATIN CAPSULES 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate.
  • a mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient.
  • the capsules are washed and dried.
  • a large number of tablets are prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose.
  • Appropriate coatings may be applied to increase palatability or delay absorption.
  • a parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol. The solution is made to volume with water for injection and sterilized.
  • aqueous suspension is prepared for oral administration so that each 5 milliliters contain 100 milligrams of finely divided active ingredient, 100 milligrams of sodium carboxymethyl cellulose, 5 milligrams of sodium benzoate, 1.0 grams of sorbitol solution, U.S.P., and 0.025 milliliters of vanillin.
  • the same dosage forms can generally be used when the compounds of this invention are administered stepwise or in conjunction with another therapeutic agent.
  • the dosage form and administration route should be selected depending on the compatibility of the combined drugs.
  • coadministration is understood to include the administration of the two agents concomitantly or sequentially, or alternatively as a fixed dose combination of the two active components.
  • Carboxylic acid ii can be activated for acylation with a reagent such as N,N'-dicyclohexylcarbodiimide, l-(3- dimethylaminopropyl)-3-ethylcarbodiimide, 1,1 '-carbonyldiimidazole, or bis(2-oxo-3- oxazolidinyl)phosphinic chloride in the presence of a suitable base (if necessary) such as triethylamine, N,N-diisopropylethylamine, or sodium bicarbonate in a solvent such as 1,2-dichloroethane, toluene, xylenes, N,N-dimethylformamide or N-methyl pynolidinone.
  • a suitable base if necessary
  • a 2-(amino)aryl N-hydroxyamidine of general structure iii can then be added which results in the formation of an acyl N-hydroxyamidine iv.
  • This intermediate can be isolated using methods known to those skilled in the art (e.g., crystallization, silica gel chromatography, HPLC) and in a subsequent step, cyclized/dehydrated by warming iv in a suitable solvent (e.g., 1,2-dichloroethane, toluene, xylenes, N,N-dimethylformamide or N-methyl pynolidinone) to give a 1,2,4- oxadiazole of structure i.
  • a suitable solvent e.g., 1,2-dichloroethane, toluene, xylenes, N,N-dimethylformamide or N-methyl pynolidinone
  • Conversion of iii to iv may require added base, in which case reagents such as pyridine, N,N-diisopropylethylarnine or tetrabutylammonium fluoride can be used. It may be more convenient or desirable to not isolate N- hydroxyamidine iv, in which case the teansformation of ii to i can be carried out as a continuous process.
  • reagents such as pyridine, N,N-diisopropylethylarnine or tetrabutylammonium fluoride
  • acylating agents other than activated carboxylic acid ii it possible to use acylating agents other than activated carboxylic acid ii to give compounds i.
  • a carboxylic acid chloride, carboxylic acid anhydride, carboxamide or carbonitrile in the place of carboxylic acid ii and an acyl activating agent to prepare 1,2,4-oxadiazole compounds i as described above.
  • Methods to prepare 1,2,4-oxadiazoles using these other acylating agents as well as other methods pertinent to the present invention are known to those skilled in the art and have been reviewed in the literature (see, Clapp, L.B., "1,2,3- and 1,2,4-Oxadiazoles", pp. 366-91 in Comprehensive Heterocyclic Chemistry, Volume 6, Potts, K. T., Editor, Pergamon Press, 1984).
  • a second method that can be used to prepare the compounds of the general stracture i in the present invention is shown in Scheme 2.
  • Carboxylic acid ii is activated as described in Scheme 1 and used to acylate a 2-(substituted)aryl N- hydroxyamidine v in which the functional group X is a leaving group such as fluoro, chloro, bromo, iodo, cyano, alkylsulfonyloxy or arylsulfonyloxy.
  • Conversion to compound iv and ring closure to give i is effected using the methods described above.
  • Displacement of the leaving group X is carried out by treating vi with ammonia, an alkylamine or a dialkylamine in a suitable solvent (e.g., methanol, ethanol, N,N- dimethylformamide, dimethylsulfoxide) at or above ambient temperature to give 1,2,4-oxadiazole i.
  • a suitable solvent e.g., methanol, ethanol, N,N- dimethylformamide, dimethylsulfoxide
  • vi can be treated with N-methylformamide in the presence of diethanolamine at elevated temperature to give compounds i, where -NR1R2 is - NHCH3.
  • R3-R8 can be manipulated in compounds i. Examples of this include (but are not limited to): 1) if one or more of R3-R8 is -OH, treatment of i with an alkyl halide or alkyl sulfonate ester in the presence of an appropriate base (e.g., N,N-diisopropylethylamine, triethylamine, pyridine, sodium carbonate) in a suitable solvent (methylene chloride, acetonitrile, toluene, N,N-dimethylformamide) at or above ambient temperature can give compounds i in which one or more of R3-R8 is alkoxy; 2) if one or more of R3- R8 is -CI, -Br, -I, or -OSO2CF3, treatment of i with an aryl boronic acid and a suitable base (sodium hydroxide, potassium bicarbonate) in the presence of
  • any of groups R1-R8 may have asymmetric centers, in which case the individual stereoisomers of i can obtained by methods known to those skilled in the art which include (but are not limited to): stereospecific synthesis, resolution of salts of i or any of the intermediates used in its preparation with enantiopure acids or bases, resolution of i or any of the intermediates used in its preparation by HPLC employing enantiopure stationary phases.
  • N-HYDROXYAMIDINE 1 2-Chloro-N-hvdroxy-nicotinamidine A mixture of 2-chloro-3-pyridine-carbonitrile (5.00 g, 37 mmol), hydroxylamine hydrochloride (3.73 g, 54 mmol) and sodium bicarbonate (9.10 g, 108 mmol) were sti ⁇ ed together in CH3OH (250 ml) at 50°C for 16 h.
  • N-HYDROXYAMIDINEs 2-6 The following N-HYDROXYAMIDINE intermediates were prepared using a procedure analogous to that described for N-HYDROXYAMIDINE 1 substituting the appropriate nitrile for 2-chloro-3-pyridine-carbonitrile.
  • N-HYDROXYAMIDINE 7 2-(N-Methylamino)-N-hydroxy-nicotinamidine A mixture of 10 g (72 mmol) 2-chloro-3-pyridine-carbonitrile, 40 mL of 40% methylamine in H2O and 20 mL of iPrOH was sti ⁇ ed at 55 °C for 1.5 h. Aqueous hydroxylamine (6.0 mL, 50 wt. % in H2O) was added and the resulting mixture was sti ⁇ ed at 55 °C for 1 h. The solution was cooled to rt.
  • N-HYDROXYAMIDINE 8 2-(Amino)-N-hydroxy-nicotinamidine
  • N-HYDROXYAMIDINE 9 3-(N-Methylamino)-pyrazine-2-(N-hydroxyamidine) Step A: 2-(N-Methylamino)-3-cyanopyrazine
  • Step B 3-(N-Methylamino)-pyrazine-2-(N-hydroxyamidine)
  • N-HYDROXYAMIDINE 10 2-(N-Methylamino)-5-fluoro-N-hvdroxynicotinamidine Step A: 2,6-Dichloro-5-fluoronicotinamide
  • Step B 2-Chloro-5-fluoronicotinamide Under a N 2 atmosphere, 2,6-dichloro-5-fluoronicotinamide (500 mg,
  • Step C 4-(Cyclobutyldifluoromethyl)benzoic acid A solution of360 mg (1.4 mmol) of ethyl 4-
  • CARBOXYLIC ACID 7 3 Trifluoromethyl-4-(2-(S)-butoxy)benzoic acid
  • Step A 3-Trifluoromethyl-4-(2-(S)-butoxy)benzonitrile
  • a solution of 1.1 g (5.9 mmol) of 4-fluoro-3 - trifluoromethylbenzonitrile and 485 mg (6.5 mmol) of (S)-(+)-2-butanol in 10 mL of THF at-10°C was treated with 235 mg (5.9 mmol) of sodium hydride. The resulting mixture was stined cold for 2 h, then quenched with 10 mL of H2O.
  • Step B 3-Trifluoromethyl-4-(l-(S)-methyl-2,2,2-trifluoroethoxy)benzoic acid
  • the title compound was prepared using procedures analogous to those described for CARBOXYLIC ACID 7 substituting 1 -(S)-mefhyl-2,2,2-trifluoroethanol (from Step A) for (S)-2-butanol in CARBOXYLIC ACID 7, Step A.
  • the enantiomeric purity of the title compound was determined by converting it to the conesponding methyl ester (excess 2.0 M trimethylsilyldiazomethane solution in cyclohexane, THF/MeOH, 5 min) and assaying by HPLC.
  • Step B 4-(2-(S)-Butoxy-2-fluoro-benzoic acid
  • Step B 5-(2-Methyl-l-oxopropyl)-2-bromopyridine
  • Step C 5-(2-Methyl-l-oxopropyl)pyridine-2-carbonitrile
  • CARBOXYLIC ACID 22 5-( 1 , 1 -Difluoro-2-methylpropyl pyridine-2-carboxylic acid
  • Step B (S)-3-(4-Bromophenyl)-l,l-difluorocyclopentane
  • Step B 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2-methylpropyl)phenyl)-l,2,4- oxadiazole
  • EXAMPLES 2-9 The following were prepared using procedures analogous to those described in EXAMPLE 1 substituting 4-(cyclohexyl)benzoic acid for 4-(2- methylpropyl)benzoic acid and the appropriate N-HYDROXYAMIDINE for N- HYDROXYAMIDINE 1 in Step A and the appropriate amine for N- methylformamide in Step B.
  • EXAMPLE 19 3 -(2-(N-Methylamino)pyridin-3 -yl -5-(4-(2.2-difluoropropyl phenvD- 1.2.4- oxadiazole
  • EXAMPLES 20-46 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl) benzoic acid and the appropriate N-HYDROXYAMIDINE for N- HYDROXYAMIDINE 1.
  • Step B 3-(2-(Benzotriazol-l-yloxy) ⁇ yridin-3-yl)-5-(5-(2- methylpropyl)pyridin-2-yl)- 1 ,2,4-oxadiazole
  • Step C 3-(2-(N-Methylamino)pyridin-3-yl)-5-(5-(2-methylpropyl)pyridin-2- yl)- 1,2,4-oxadiazole
  • Step A 3-(2-(Chloro)pyridin-3-yl)-5-(4-hydroxyphenyl)- 1 ,2,4-oxadiazole
  • Step B 3-(2-(Chloro)pyridin-3-yl)-5-(4-(2,2,2-trifluoroethoxy)phenyl)-l,2,4- oxadiazole
  • Step C 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2,2,2-trifluoroethoxy)phenyl)- 1,2,4-oxadiazole
  • Step B 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2-fluoro-l-fluoromethyl)ethoxy-3- trifluoromethylphenyl) - 1 ,2,4-oxadiazole
  • EXAMPLES 73-80 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
  • EXAMPLES 88-90 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE l and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
  • HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
  • EXAMPLES 103-106 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
  • EXAMPLE 107 3-(2-Amino-5-fluoropyridin-3-yl)-5-(3-trifluoromethyl-4-(l,l.l-trifluoro-2-(S)- propyloxy))phenyl)- 1 ,2,4-oxadiazole
  • EDC-HC1 34 mg, 0.176 mmol
  • the resultant solution was added to a mixture of N-HYDROXYAMIDINE 11 and acetonifrile (1.0 mL) in a sealed tube and heated to 40 °C.
  • EXAMPLE 108 3-(2-(TS-Methylamino)-5-fluoropyridin-3-yl)-5-(3-trifluoromethyl-4-(l,l,l-trifluoro- 2-(S)-propyloxy) phenyl)- 1 ,2,4-oxadiazole
  • the title compound was prepared using a procedure analogous to that described for EXAMPLE 107 substituting N-HYDROXYAMIDINE 10 for N-
  • SlPi/Edgl, SlP3,/Edg3, SlP2/Edg5, SlP4/Edg6 or SIP5 /Edg8 activity of the compounds of the present invention can be evaluated using the following assays:
  • Reaction products were extracted with butanol and 33p_ S phingosine-l- phosphate was purified by HPLC.
  • Cells expressing EDG/S1P receptors were harvested with enzyme-free dissociation solution (Specialty Media, Lavallette, NJ). They were washed once in cold PBS and suspended in binding assay buffer consisting of 50 mM HEPES-Na, pH 7.5, 5mM MgCl2, lmM CaCl2, and 0.5% fatty acid-free BSA.
  • 33p- sp hfngosine-l- phosphate was sonicated with 0.1 nM sphingosine- 1-phosphate in binding assay buffer; 100 ⁇ l of the ligand mixture was added to 100 ⁇ l cells (1 x 106 cells/ml) in a 96 well microtiter dish. Binding was performed for 60 min at room temperature with gentle mixing. Cells were then collected onto GF/B filter plates with a Packard Filtermate Universal Harvester. After drying the filter plates for 30 min, 40 ⁇ l of Microscint 20 was added to each well and binding was measured on a Wallac
  • Non-specific binding was defined as the amount of radioactivity remaining in the presence of 0.5 ⁇ M cold sphingosine- 1-phosphate.
  • ligand binding assays were performed on membranes prepared from cells expressing Edg/SIP receptors.
  • Cells were harvested with enzyme-free dissociation solution and washed once in cold PBS.
  • Cells were disrupted by homogenization in ice cold 20 mM HEPES pH 7.4, 10 mM EDTA using a Kinematica polytron (setting 5, for 10 seconds).
  • Homogenates were centrifuged at 48,000 x g for 15 min at 40C and the pellet was suspended in 20 mM HEPES pH 7.4, 0.1 mM EDTA.
  • the final pellet was suspended in 20 mM HEPES pH 7.4, 100 mM NaCl, 10 mM MgCl2- Ligand binding assays were performed as described above, using 0.5 to 2 ⁇ g of membrane protein.
  • Agonists and antagonists of Edg/SIP receptors can be identified in the 33p-sphingosine- 1-phosphate binding assay.
  • Compounds diluted in DMSO, methanol, or other solvent, were mixed with probe containing 33p_sphingosine-l- phosphate and binding assay buffer in microtiter dishes.
  • Membranes prepared from cells expressing Edg/SIP receptors were added, and binding to 3p_sphingosine-l- phosphate was performed as described. Determination of the amount of binding in the presence of varying concentrations of compound and analysis of the data by nonlinear regression software such as MRLCalc (Merck Research Laboratories) or PRISM (GraphPad Software) was used to measure the affinity of compounds for the receptor.
  • Selectivity of compounds for Edg/SIP receptors was determined by measuring the level of 33p_sphingosine- 1-phosphate binding in the presence of the compound using membranes prepared from cells transfected with each respective receptor (SlPi/Edgl, SlP3/Edg3, SlP2/Edg5, SlP4/Edg6, SlPs/Edg8).
  • Agonists and antagonists of SlP/Edg receptors can be discriminated in the 35s-GTP ⁇ S binding assay.
  • Compounds diluted in DMSO, methanol, or other solvent, were added to microtiter dishes to provide final assay concentrations of 0.01 nM to 10 ⁇ M.
  • Membranes prepared from cells expressing SlP/Edg receptors were added, and binding to 5s-GTP ⁇ S was performed as described. When assayed in the absence of the natural ligand or other known agonist, compounds that stimulate 35 s- GTP ⁇ S binding above the endogenous level were considered agonists, while compounds that inhibit the endogenous level of 35s-GTP ⁇ S binding were considered inverse agonists.
  • Antagonists were detected in a 35s-GTP ⁇ S binding assay in the presence of a sub-maximal level of natural ligand or known SlP/Edg receptor agonist, where the compounds reduced the level of 35s-GTP ⁇ S binding. Determination of the amount of binding in the presence of varying concentrations of compound was used to measure the potency of compounds as agonists, inverse agonists, or antagonists of SlP/Edg receptors. To evaluate agonists, percent stimulation over basal was calculated as binding in the presence of compound divided by binding in the absence of ligand, multiplied by 100.
  • Dose response curves were plotted using a non-linear regression curve fitting program MRLCalc (Merck Research Laboratories), and EC50 values were defined to be the concentration of agonist required to give 50% of its own maximal stimulation.
  • Selectivity of compounds for SlP/Edg receptors was determined by measuring the level of 35S-GTP ⁇ S binding in the presence of compound using membranes prepared from cells transfected with each respective receptor.
  • a 96-well ligand plate was prepared by diluting sphingosine- 1- phosphate or other agonists into 200 ⁇ l of assay buffer to give a concentration that was 2-fold the final test concentration.
  • the ligand plate and the cell plate were loaded into the FLIPR instrument for analysis. Plates were equilibrated to 37°C.
  • the assay was initiated by transfening an equal volume of ligand to the cell plate and the calcium flux was recorded over a 3 min interval. Cellular response was quantitated as area (sum) or maximal peak height (max).
  • Antagonists were evaluated in the absence of natural ligand by dilution of compounds into the appropriate solvent and transfer to the Fluo-4 labeled cells.
  • Antagonists were evaluated by pretreating Fluo-4 labeled cells with varying concentrations of compounds for 15 min prior to the initiation of calcium flux by addition of the natural ligand or other SlP/Edg receptor agonist.
  • SlPi/Edgl SlP3/Edg3, SlP2/Edg5, SlP4/Edg6 or SlP5/Edg8.
  • RACE PCR cloning technique Frohman, et al., 1988, Proc. Natl. Acad. Sci. USA 85: 8998-9002.
  • 5' and/or 3' RACE may be performed to generate a full-length cDNA sequence; (2) direct functional expression of the Edg/SIP cDNA following the construction of an SlP/Edg-containing cDNA library in an appropriate expression vector system; (3) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a labeled degenerate oligonucleotide probe designed from the amino acid sequence of the SlP/Edg protein; (4) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA encoding the SlP/Edg protein.
  • This partial cDNA is obtained by the specific PCR amplification of SlP/Edg DNA fragments through the design of degenerate oligonucleotide primers from the amino acid sequence known for other proteins which are related to the SlP/Edg protein; (5) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA or oligonucleotide with homology to a mammalian SlP/Edg protein.
  • This strategy may also involve using gene-specific oligonucleotide primers for PCR amplification of SlP/Edg cDNA; or (6) designing 5' and 3' gene specific oligonucleotides using the SlP/Edg nucleotide sequence as a template so that either the full-length cDNA may be generated by known RACE techniques, or a portion of the coding region may be generated by these same known RACE techniques to generate and isolate a portion of the coding region to use as a probe to screen one of numerous types of cDNA and/or genomic libraries in order to isolate a full-length version of the nucleotide sequence encoding SlP/Edg.
  • libraries as well as libraries constructed from other cell types-or species types, may be useful for isolating an SlP/Edg-encoding DNA or an SlP/Edg homologue.
  • Other types of libraries include, but are not limited to, cDNA libraries derived from other cells.
  • suitable cDNA libraries may be prepared from cells or cell lines which have SlP/Edg activity.
  • the selection of cells or cell lines for use in preparing a cDNA library to isolate a cDNA encoding SlP/Edg may be done by first measuring cell-associated SlP/Edg activity using any known assay available for such a purpose.
  • cDNA libraries can be performed by standard techniques well known in the art. Well known cDNA library construction techniques can be found for example, in Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Complementary DNA libraries may also be obtained from numerous commercial sources, including but not limited to Clontech Laboratories, Inc. and Stratagene.
  • An expression vector containing DNA encoding an SlP/Edg-like protein may be used for expression of S lP/Edg in a recombinant host cell.
  • a recombinant host cell can be cultured under suitable conditions to produce SlP/Edg or a biologically equivalent form.
  • Expression vectors may include, but are not limited to, cloning vectors, modified cloning vectors, specifically designed plasmids or viruses. Commercially available mammalian expression vectors may be suitable for recombinant S 1 P/Edg expression.
  • Recombinant host cells may be prokaryotic or eukaryotic, including but not limited to, bacteria such as E. coli, fungal cells such as yeast, mammalian cells including, but not limited to, cell lines of bovine, porcine, monkey and rodent origin; and insect cells including but not limited to Drosophila and silkworm derived cell lines.
  • bacteria such as E. coli
  • fungal cells such as yeast
  • mammalian cells including, but not limited to, cell lines of bovine, porcine, monkey and rodent origin
  • insect cells including but not limited to Drosophila and silkworm derived cell lines.
  • SlP/Edg receptors The nucleotide sequences for the various SlP/Edg receptors are known in the art. See, for example, the following: SlPj/Edgl Human
  • EDG6 a novel G-protein- coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue. Genomics 53: 164-169, hereby incorporated by reference in its entirety. WO 98/48016, published October 29, 1998, hereby incorporated by reference in its entirety.
  • Rats were instrumented with femoral arterial and venous catheters for measurement of arterial pressure and intravenous compound administration, respectively. Animals were anesthetized with Nembutal (55 mg/kg, ip). Blood pressure and heart rate were recorded on the Gould Po-Ne-Mah data acquisition system. Heart rate was derived from the arterial pulse wave. Following an acclimation period, a baseline reading was taken (approximately 20 minutes) and the data averaged. Compound was administered intravenously (either bolus injection of approximately 5 seconds or infusion of 15 minutes duration), and data were recorded every 1 minute for 60 minutes post compound administration.
  • Nembutal 55 mg/kg, ip
  • Heart rate was derived from the arterial pulse wave. Following an acclimation period, a baseline reading was taken (approximately 20 minutes) and the data averaged.
  • Compound was administered intravenously (either bolus injection of approximately 5 seconds or infusion of 15 minutes duration), and data were recorded every 1 minute for 60 minutes post
  • Data are calculated as either the peak change in heart rate or mean arterial pressure or are calculated as the area under the curve for changes in heart rate or blood pressure versus time. Data are expressed as mean + SEM. A one-tailed Student's paired t-test is used for statistical comparison to baseline values and considered significant at p ⁇ 0.05.
  • a single mouse is dosed intravenously (tail vein) with 0.1 ml of test compound dissolved in a non-toxic vehicle and is observed for signs of toxicity. Severe signs may include death, seizure, paralysis or unconciousness. Milder signs are also noted and may include ataxia, labored breathing, ruffling or reduced activity relative to normal.
  • the dosing solution is diluted in the same vehicle. The diluted dose is administered in the same fashion to a second mouse and is likewise observed for signs. The process is repeated until a dose is reached that produces no signs. This is considered the estimated no-effect level. An additional mouse is dosed at this level to confirm the absence of signs.
  • 0.5 ml of blood is withdrawn via direct cardiac puncture, blood is immediately stabilized with EDTA and hematology is evaluated using a clinical hematology autoanalyzer calibrated for performing murine differential counts (H2000, CARESIDE, Culver City CA).
  • H2000, CARESIDE, Culver City CA a clinical hematology autoanalyzer calibrated for performing murine differential counts
  • Reduction in lymphocytes by test treatment is established by comparison of hematological parameters of three mice versus three vehicle treated mice.
  • the dose used for this evaluation is determined by tolerability using a modification of the dilution method above. For this purpose, no-effect is desirable, mild effects are acceptable and severely toxic doses are serially diluted to levels that produce only mild effects.
  • the examples disclosed herein have utility as immunoregulatory agents as demonstrated by their activity as potent and selective agonists of the SlPi/Edgl receptor over the S1PR3/Edg3 receptor as measured in the assays described above.
  • the examples disclosed herein possess a selectivity for the SlPi/Edgl receptor over the S1PR3/Edg3 receptor of more than 100 fold as measured by the ratio of EC50 for the SlPi/Edgl receptor to the EC50 for the SlP3/Edg3 receptor as evaluated in the 35s-GTP ⁇ S binding assay described above and possess an EC50 for binding to the SlPi/Edgl receptor of less than 50 nM as evaluated by the 35s-GTP ⁇ S binding assay described above.

Abstract

The present invention encompasses compounds of Formula (I): as well as the pharmaceutically acceptable salts thereof. The compounds are useful for treating immune mediated diseases and conditions, such as bone marrow, organ and tissue transplant rejection. Pharmaceutical compositions and methods of use are included.

Description

TITLE OF THE INVENTION
3-(2-AMTNO-l-AZACYCLYL)-5-ARYL-l,2,4-OXADIAZOLES AS SIP RECEPTOR AGONISTS
BACKGROUND OF THE INVENTION
The present invention is related to compounds that are SlPi/Edgl receptor agonists and thus have immunosuppressive activities by modulating leukocyte trafficking, sequestering lymphocytes in secondary lymphoid tissues, and interfering with celkcell interactions required for an efficient immune response. The invention is also directed to pharmaceutical compositions containing such compounds and methods of treatment or prevention.
Immunosuppressive agents have been shown to be useful in a wide variety of autoimmune and chronic inflammatory diseases, including systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis and other disorders such as Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy, atopic dermatitis and asthma. They have also proved useful as part of chemotherapeutic regimens for the treatment of cancers, lymphomas and leukemias. Although the underlying pathogenesis of each of these conditions may be quite different, they have in common the appearance of a variety of autoantibodies and/or self-reactive lymphocytes. Such self-reactivity may be due, in part, to a loss of the homeostatic controls under which the normal immune system operates. Similarly, following a bone-maπow or an organ transplantation, the host lymphocytes recognize the foreign tissue antigens and begin to produce both cellular and humoral responses including antibodies, cytokines and cytotoxic lymphocytes which lead to graft rejection.
One end result of an autoimmune or a rejection process is tissue destruction caused by inflammatory cells and the mediators they release. Anti- inflammatory agents such as NSAIDs act principally by blocking the effect or secretion of these mediators but do nothing to modify the immunologic basis of the disease. On the other hand, cytotoxic agents, such as cyclophosphamide, act in such a nonspecific fashion that both the normal and autoimmune responses are shut off. Indeed, patients treated with such nonspecific immunosuppressive agents are as likely to succumb to infection as they are to their autoimmune disease.
Cyclosporin A is a drag used to prevent rejection of transplanted organs. FK-506 is another drug approved for the prevention of transplant organ rejection, and in particular, liver transplantation. Cyclosporin A and FK-506 act by inhibiting the body's immune system from mobilizing its vast arsenal of natural protecting agents to reject the transplant's foreign protein. Cyclosporin A was approved for the treatment of severe psoriasis and has been approved by European regulatory agencies for the treatment of atopic dermatitis. Though they are effective in delaying or suppressing transplant rejection, Cyclosporin A and FK-506 are known to cause several undesirable side effects including nephrotoxicity, neurotoxicity, and gastrointestinal discomfort. Therefore, an immunosuppressant without these side effects still remains to be developed and would be highly desirable. The immunosuppressive compound FTY720 is a lymphocyte sequestration agent cunently in clinical trials. FTY720 is metabolized in mammals to a compound that is a potent agonist of sphingosine 1 -phosphate receptors. Agonism of sphingosine 1 -phosphate receptors modulates leukocyte trafficking, induces the sequestration of lymphocytes (T-cells and B-cells) in lymph nodes and Peyer's patches without lymphodepletion, and disrapts splenic architecture, thereby interfering with T cell dependent and independent antibody responses. Such immunosuppression is desirable to prevent rejection after organ transplantation and in the treatment of autoimmune disorders.
Sphingosine 1 -phosphate is a bioactive sphingolipid metabolite that is secreted by hematopoietic cells and stored and released from activated platelets. Yatomi, Y., T. Ohmori, G. Rile, F. Kazama, H. Okamoto, T. Sano, K. Satoh, S. Kume, G. Tigyi, Y. Igarashi, and Y. Ozaki. 2000. Blood. 96:3431-8. It acts as an agonist on a family of G protein-coupled receptors to regulate cell proliferation, differentiation, survival, and motility. Fukushima, N., I. Ishii, J. J.A. Contos, J.A. Weiner, and J. Chun. 2001. Lysophospholipid receptors. Annu. Rev. Pharmacol.
Toxicol. 41:507-34; Hla, T., M.-J. Lee, N. Ancellin, J.H. Paik, and MJ. Kluk. 2001. Lysophospholipids - Receptor revelations. Science. 294:1875-1878; Spiegel, S., and S. Milstien. 2000. Functions of a new family of sphingosine- 1 -phosphate receptors. Biochim. Biophys. Ada. 1484:107-16; Pyne, S., and N. Pyne. 2000. Sphingosine 1- phosphate signalling via the endothelial differentiation gene family of G-protein coupled receptors. Pharm. & Therapeutics. 88:115-131. Five sphingosine 1- phosphate receptors have been identified (SlPi, S1P2, SIP3, SIP4, and SIP5, also known as endothelial differentiation genes Edgl, Edg5, Edg3, Edg6, Edg8), that have widespread cellular and tissue distribution and are well conserved in human and rodent species (see Table). Binding to SIP receptors elicits signal transduction through Gq-, Gi/o, G12-, G13-, and Rho-dependent pathways. Ligand-induced activation of SIP 1 and SIP3 has been shown to promote angiogenesis, chemotaxis, and adherens junction assembly through Rac- and Rho-, see Lee, M.-J., S. Thangada, K.P. Claffey, N. Ancellin, CH. Liu, M. Kluk, M. Volpi, R.I. Sha'afi, and T. Hla. 1999. Cell. 99:301-12, whereas agonism of SIP2 promotes neurite retraction, see
Van Brooklyn, J.R., Z. Tu, L.C. Edsall, R.R. Schmidt, and S. Spiegel. 1999. J. Biol. Chem. 274:4626-4632, and inhibits chemotaxis by blocking Rac activation, see Okamoto, H., N. Takuwa, T. Yokomizo, N. Sugimoto, S. Sakurada, H. Shigematsu, and Y. Takuwa. 2000. Mol. Cell. Biol. 20:9247-9261. SIP4 is localized to hematopoietic cells and tissues, see Graeler, M.H., G. Bernhardt, and M. Lipp. 1999. Curr. Top. Microbiol. Immunol. 246:131-6, whereas SIP5 is primarily a neuronal receptor with some expression in lymphoid tissue, see hn, D.S., C.E. Heise, N. Ancellin, B.F. O'Dowd, G.J. Shei, R.P. Heavens, M.R. Rigby, T. Hla, S. Mandala, G. McAllister, S. George, and K.R. Lynch. 2000. J. Biol. C em. 275:14281-6. Administration of sphingosine 1 -phosphate to animals induces systemic sequestration of peripheral blood lymphocytes into secondary lymphoid organs, thus resulting in therapeutically useful immunosuppression, see Mandala, S., R. Hajdu, J. Bergstiom, E. Quackenbush, J. Xie, J. MiUigan, R. Thornton, G.-J. Shei, D. Card, C. Keohane, M. Rosenbach, J. Hale, C.L. Lynch, K. Rupprecht, W. Parsons, H. Rosen. 2002. Science. 296:346-349. However, sphingosine 1-phosphate also has cardiovascular and bronchoconstrictor effects that limit its utility as a therapeutic agent. Intravenous administration of sphingosine 1-phosphate decreases the heart rate, ventricular contraction and blood pressure in rats, see Sugiyama, A., N.N. Aye, Y. Yatomi, Y. Ozaki, and K. Hashimoto. 2000. Jpn. J. Pharmacol. 82:338-342. In human airway smooth muscle cells, sphingosine 1-phosphate modulates contraction, cell growth and cytokine production that promote bronchoconstriction, airway inflammation and remodeling in asthma, see Ammit, A.J., A.T. Hastie, L. C. Edsall, R.K. Hoffman, Y. Amrani, V.P. Krymskaya, S.A. Kane, S.P. Peters, R.B. Perm, S. Spiegel, RA. Panettieri. Jr. 2001, FASEB J. 15:1212-1214. The undesirable effects of sphingosine 1-phosphate are associated with its non-selective, potent agonist activity on all SIP receptors.
The present invention encompasses compounds which are agonists of the SlPi/Edgl receptor having selectivity over the SlP3/Edg3 receptor. An SlPl/Edgl receptor selective agonist has advantages over cuπent therapies and extends the therapeutic window of lymphocyte sequestration agents, allowing better tolerability with higher dosing and thus improving efficacy as monotherapy.
While the main use for immunosuppressants is in treating bone maπow, organ and transplant rejection, other uses for such compounds include the treatment of arthritis, in particular, rheumatoid arthritis, insulin and non-insulin dependent diabetes, multiple sclerosis, psoriasis, inflammatory bowel disease, Crohn's disease, lupus erythematosis and the like.
Thus, the present invention is focused on providing immunosuppressant compounds that are safer and more effective than prior compounds. These and other objects will be apparent to those of ordinary skill in the art from the description contained herein.
Figure imgf000005_0001
Figure imgf000006_0003
SUMMARY OF THE INVENTION
The present invention encompasses compounds of Formula I:
as well as the pharmaceutically acceptable salts thereof. The compounds are useful for treating immune mediated diseases and conditions, such as bone manow, organ and tissue transplant rejection. Pharmaceutical compositions and methods of use are included.
DETAILED DESCRIPTION OF THE INVENTION The present invention encompasses compounds represented by
Formula I:
Figure imgf000006_0002
or a pharmaceutically acceptable salt thereof, wherein:
A is C-R3 orN,
D is C-R orN,
E is C-R6 orN and
G is C-R7 orN,
with the proviso that at least one of A, D, E and G is not N;
X, Y and Z are independently selected from the group consisting of: N and C-R8, with the proviso that at least one of X, Y and Z is not N;
Rl and R2 are each independently selected from the group consisting of:
(1) hydogen and
(2) Ci-6alkyl, optionally substituted with 1 to 3 halo groups, or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form a 3- to 6-membered saturated monocyclic ring;
R3, R4, R6 and R7 are each independently selected from the group consisting of:
(1) hydrogen,
(2) halo (3) cyano, and
(4) Ci-4alkyl or Ci-4alkoxy, each optionally substituted with 1 to
3 halo groups;
R5 is selected from the group consisting of: (1) Ci-6alkyl,
(2) C2-6alkenyl,
(3) C2-6alkynyl,
(4) C3-6cycloalkyl, (5) Cι_6alkoxy,
(6) C3-6cycloalkoxy,
(7) Ci-βacyl,
(8) halo, (9) aryl and
(10) HET, wherein groups (1) to (7) above are optionally substituted with from one up to the maximum number of substituable positions with halo, and
groups (9) and (10) above are optionally substituted with 1 to 3 substituents independently selected from the group consisting of:
(a) halo, and
(b) Ci-4alkyl or Ci-4alkoxy, each optionally substituted with oxo, hydroxy or 1 to 3 halo groups,
or R4 and R5 may be joined together with the atoms to which they are attached to form a 5 or 6-membered monocyclic ring, optionally containing 1 to 3 heteratoms selected from O, S and NR8, said ring optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, Ci-4alkyl and Ci-4alkoxy, said Ci-4alkyl or Ci-4alkoxy optionally substituted with 1 to 3 halo groups;
each R8 is independently selected from the group consisting of: hydrogen, halo and Cl-4alkyl, wherein said Cl-4alkyl is optionally substituted with 1 to 3 halo groups; and
HET is selected from the group consisting of: benzimidazolyl, benzofuranyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pyπolyl, quinazolinyl, quinolyl, quinoxalinyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pynolidinyl, mo holinyl, thiomorpholinyl, dihydrobenzimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropynolyl, dihydroquinolinyl, dihydroteteazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetiahydrofuranyl, and tetrahydrothienyl.
An embodiment of the invention encompasses a compound of Formula
I wherein:
A is N,
D is C-R4,
E is C-R6 and
G is C-R7.
Another embodiment of the invention encompasses a compound of
Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7. Another embodiment of the invention encompasses a compound of Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7; and
X, Y and Z are C-R8.
Another embodiment of the invention encompasses a compound of Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7; and
R3, R6 and R7 are hydrogen. Within this embodiment is encompassed a compound of Formula I wherein R is trifluoromethyl or cyano.
Another embodiment of the invention encompasses a compound of Formula I wherein:
A is C-R3,
D is C-R4, E is C-R6 and
G is C-R7; and
Rl and R2 are each independently selected from the group consisting of hydrogen, methyl and ethyl.
Another embodiment of the invention encompasses a compound of Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7; and
R5 is selected from the group consisting of:
(1) C2-6alkyl,
(2) C3-6cycloalkyl,
(3) C2-6alkoxy,
(4) C3-6cycloalkoxy, and (5) C3-6acyl, wherein groups (1) to (5) above are optionally substituted with 1 to 5 fluoro groups. Within this embodiment is encompassed a compound of Formula I wherein R5 is C2- βalkoxy, optionally substituted with 1 to 5 fluoro groups.
Another embodiment of the invention encompasses a compound of
Formula I wherein:
A is C-R3, D is C-R4,
E is C-R6 and
G is C-R7; and
R5 is selected from the group consisting of:
(1) phenyl, optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, methyl, methoxy and hydroxymethyl,
(2) oxadiazolyl,
(3) oxazolyl,
(4) furanyl and (5) thienyl.
Another embodiment of the invention encompasses a compound of Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7; and
X is N and Y and Z are both C-R8.
Another embodiment of the invention encompasses a compound of
Formula I wherein:
A is C-R3, D is C-R4,
E is C-R6 and
G is C-R7; and
wherein X and Z are both C-R.8 and Y is N.
Another embodiment of the invention encompasses a compound of
Formula I wherein:
A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7;
Rl and R2 are each independently selected from the group consisting of: hydrogen and methyl,
R3, R6 and R are hydrogen,
R4 is trifluoromethyl or cyano, and
R5 is C2-6alkoxy, optionally substituted with 1 to 5 fluoro groups. Within this embodiment is encompassed an invention wherein R5 is selected from 2,2,2- trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy. Also within this embodiment is encompassed an invention wherein R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy, X, Y and Z are C-R§ and each R8 is independently selected from hydrogen, methyl and halo. Also within this embodiment is encompassed an invention wherein R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy, X is N and Y and Z are both C-R8 and each R8 is independently selected from hydrogen, methyl and halo. Also within this embodiment is encompassed an invention wherein R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy, X and Z are both C-R8 and Y is N and each Ro" is independently selected from hydrogen, methyl and halo.
Exemplifying the invention are the following compounds:
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
or a pharmaceutically acceptable salt of any of the above.
The invention also encompasses a method of treating an immunoregulatory abnormality in a mammalian patient in need of such treatment comprising administering to said patient a compound of Formula I in an amount that is effective for treating said immunoregulatory abnormality.
Within this embodiment is encompassed the above method wherein the immunoregulatory abnormality is an autoimmune or chronic inflammatory disease selected from the group consisting of: systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy and asthma.
Also within this embodiment is encompassed the above method wherein the immunoregulatory abnormality is bone manow or organ transplant rejection or graft-versus-host disease.
Also within this embodiment is encompassed the above method wherein the immunoregulatory abnormality is selected from the group consisting of: transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, post-infectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous deπnatitis, sebonhoeic deπnatitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa, urticaria, angioedemas, vasculitis, eiythema, cutaneous eosinophilia, lupus erythematosus, acne, alopecia areata, keratoconjunctivitis, vernal conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis comeae, comeal leukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' opthalmopathy, Vogt-Koyanagi-Harada syndrome, sarcoidosis, pollen allergies, reversible obstructive airway disease, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, chronic or inveterate asthma, late asthma and airway hyper-responsiveness, bronchitis, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic bowel diseases, inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal bums, coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, ulcerative colitis, migraine, rhinitis, eczema, interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome, diabetic nephropathy, multiple myositis, Guillain-Baπe syndrome, Meniere's disease, polyneuritis, multiple neuritis, mononeuritis, radiculopathy, hyperthyroidism, Basedow's disease, pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia, anerythroplasia, osteoporosis, sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photoallergic sensitivity, cutaneous T cell lymphoma, arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, myocardosis, scleroderma, Wegener's granuloma, Sjogren's syndrome, adiposis, eosinophilic fascitis, lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis, glomeralonephritis, male pattern alopecia or alopecia senilis by preventing epilation or providing hair germination and/or promoting hair generation and hair growth, muscular dystrophy, pyoderma and Sezary's syndrome, Addison's disease, ischemia-reperfusion injury of organs which occurs upon preservation, transplantation or ischemic disease, endotoxin-shock, pseudomembranous colitis, colitis caused by drug or radiation, ischemic acute renal insufficiency, chronic renal insufficiency, toxinosis caused by lung-oxygen or drugs, lung cancer, pulmonary emphysema, cataracta, siderosis, retinitis pigmentosa, senile macular degeneration, vitieal scarring, comeal alkali bu , dermatitis erythema multiforme, linear IgA ballous dermatitis and cement dermatitis, gingivitis, periodontitis, sepsis, pancreatitis, diseases caused by environmental pollution, aging, carcinogenesis, metastasis of carcinoma and hypobaropathy, disease caused by histamine or leukotriene-C4 release, Behcet's disease, autoimmune hepatitis, primary biliary cinhosis, sclerosing cholangitis, partial liver resection, acute liver necrosis, necrosis caused by toxin, viral hepatitis, shock, or anoxia, B-virus hepatitis, non- A/non-B hepatitis, cinhosis, alcoholic cinhosis, hepatic failure, fulminant hepatic failure, late-onset hepatic failure, "acute-on-chronic" liver failure, augmentation of chemotherapeutic effect, cytomegaloviras infection, HCMN infection, AIDS, cancer, senile dementia, trauma, and chronic bacterial infection.
Also within this embodiment is encompassed the above method wherein the immunoregulatory abnormality is selected from the group consisting of:
1) multiple sclerosis,
2) rheumatoid arthritis, 3) systemic lupus erythematosus,
4) psoriasis,
5) rejection of transplanted organ or tissue,
6) inflammatory bowel disease,
7) a malignancy of lymphoid origin, 8) acute and chronic lymphocytic leukemias and lymphomas and
9) insulin and non-insulin dependent diabetes. The invention also encompasses a method of suppressing the immune system in a mammalian patient in need of immunosuppression comprising administering to said patient an immunosuppressmg effective amount of a compound of Formula I.
The invention also encompasses a phaπnaceutical composition comprised of a compound of Formula I in combination with a pharmaceutically acceptable carrier.
The invention also encompasses a method of treating a respiratory disease or condition in a mammalian patient in need of such treatment comprising administering to said patient a compound of Formula I in an amount that is effective for treating said respiratory disease or condition. Within this embodiment is encompasses the above method wherein the respiratory disease or condition is selected from the group consisting of: asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia.
Also, within this embodiment is encompassed the above method wherein the patient also has a respiratory disease or condition.
Also, within this embodiment is encompassed the above method wherein the patient is also suffering from a cardiovascular disease or condition. The invention is described using the following definitions unless otherwise indicated.
When a nitrogen atom appears in a formula of the present specification, it is understood that sufficient hydrogen atoms or substituents are present to satisfy the valency of the nitrogen atom. The term "halogen" or "halo" includes F, CI, Br, and I.
The term "alkyl" means linear or branched structures and combinations thereof, having the indicated number of carbon atoms. Thus, for example, Ci-6alkyl includes methyl, ethyl, propyl, 2-propyl, s- and t-butyl, butyl, pentyl, hexyl, 1,1- dimethylethyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The term "alkoxy" means alkoxy groups of a straight, branched or cyclic configuration having the indicated number of carbon atoms. Ci-6alkoxy, for example, includes methoxy, ethoxy, propoxy, isopropoxy, and the like.
The term "alkylthio" means alkylthio groups having the indicated number of carbon atoms of a straight, branched or cyclic configuration. Ci- 6alkylthio, for example, includes methylthio, propylthio, isopropylthio, and the like.
The term "alkenyl" means linear or branched structures and combinations thereof, of the indicated number of carbon atoms, having at least one carbon-to-carbon double bond, wherein hydrogen may be replaced by an additional carbon-to-carbon double bond. C2-6alkenyl, for example, includes ethenyl, propenyl, 1-methylethenyl, butenyl and the like.
The term "alkynyl" means linear or branched structures and combinations thereof, of the indicated number of carbon atoms, having at least one carbon-to-carbon triple bond. C3_6alkynyl, for example, includes , propenyl, 1- methylethenyl, butenyl and the like.
The term "cycloalkyl" means mono-, bi- or tri-cyclic structures, optionally combined with linear or branched structures, having the indicated number of carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclopentyl, cycloheptyl, adamantyl, cyclododecylmethyl, 2-ethyl-l- bicyclo[4.4.0]decyl, cyclobutylmethyl and the like.
The term "cycloalkoxy" means cycloalkyl-O- wherein cycloalkyl is as defined above. For example, cycloalkoxy includes cyclobutoxy. The term "acyl" means an alkyl group as defined above substituted at the 1 -position with oxo. Examples include formyl, acetyl, propionyl, butyryl, valeryl and hexanoyl.
The term "aryl" is defined as a mono- or bi-cyclic aromatic ring system and includes, for example, phenyl, naphthyl, and the like. The term "aralkyl" means an alkyl group as defined above of 1 to 6 carbon atoms with an aryl group as defined above substituted for one of the alkyl hydrogen atoms, for example, benzyl and the like.
The term "aryloxy" means an aryl group as defined above attached to a molecule by an oxygen atom (aryl-O) and includes, for example, phenoxy, naphthoxy and the like.
The term "aralkoxy" means an aralkyl group as defined above attached to a molecule by an oxygen atom (aralkyl-O) and includes, for example, benzyloxy, and the like.
The term "arylthio" is defined as an aryl group as defined above attached to a molecule by an sulfur atom (aryl-S) and includes, for example, thiophenyoxy, thionaphthoxy and the like.
The term "aroyl" means an aryl group as defined above attached to a molecule by an carbonyl group (aryl-C(O)-) and includes, for example, benzoyl, naphthoyl and the like. The term "aroyloxy" means an aroyl group as defined above attached to a molecule by an oxygen atom (aroyl-O) and includes, for example, benzoyloxy or benzoxy, naphthoyloxy and the like. The term "treating" encompasses not only treating a patient to relieve the patient of the signs and symptoms of the disease or condition but also prophylactically treating an asymptomatic patient to prevent the onset or progression of the disease or condition. The term "amount effective for treating" is intended to mean that amount of a drag or pharmaceutical agent that will elicit the biological or medical response of a tissue, a system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. The term also encompasses the amount of a pharmaceutical dmg that will prevent or reduce the risk of occunence of the biological or medical event that is sought to be prevented in a tissue, a system, animal or human by a researcher, veterinarian, medical doctor or other clinician.
The invention described herein includes pharmaceutically acceptable salts and hydrates. Pharmaceutically acceptable salts include both the metallic (inorganic) salts and organic salts; a list of which is given in Remington's Pharmaceutical Sciences, 17th Edition, pg. 1418 (1985). It is well known to one skilled in the art that an appropriate salt form is chosen based on physical and chemical stability, flowability, hydroscopicity and solubility. As will be understood by those skilled in the art, pharmaceutically acceptable salts include, but are not limited to salts of inorganic acids such as hydrochloride, sulfate, phosphate, diphosphate, hydrobromide, and nitrate or salts of an organic acid such as malate, maleate, fumarate, tartiate, succinate, citrate, acetate, lactate, methanesulfonate, p- toluenesulfonate or pamoate, salicylate and stearate. Similarly pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium and ammonium (especially ammonium salts with secondary amines). Prefeπed salts of this invention for the reasons cited above include potassium, sodium, calcium and ammonium salts. Also included within the scope of this invention are crystal forms, hydrates and solvates of the compounds of Formula I.
For purposes of this Specification, "pharmaceutically acceptable hydrate" means the compounds of the instant invention crystallized with one or more molecules of water to form a hydrated form. The invention also includes the compounds falling within Formula I in the form of one or more stereoisomers, in substantially pure form or in the form of a mixture of stereoisomers. All such isomers are encompassed within the present invention. By virtue of their SlPl/Edgl agonist activity, the compounds of the present invention are immunoregulatory agents useful for treating or preventing automimmune or chronic inflammatory diseases. The compounds of the present invention are useful to suppress the immune system in instances where immunosuppression is in order, such as in bone manow, organ or transplant rejection, autoimmune and chronic inflammatory diseases, including systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy and asthma.
More particularly, the compounds of the present invention are useful to treat or prevent a disease or disorder selected from the group consisting of: transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomeralonephritis, post-infectious autoimmune diseases including rheumatic fever and post-infectious glomeralonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, seboπhoeic deπnatitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa, urticaria, angioedemas, vasculitis, erythema, cutaneous eosinophilia, lupus erythematosus, acne, alopecia areata, keratoconjunctivitis, vernal conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis corneae, comeal leukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' opthalmopathy, Vogt-Koyanagi-Harada syndrome, sarcoidosis, pollen allergies, reversible obstructive airway disease, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, chronic or inveterate asthma, late asthma and airway hyper-responsiveness, bronchitis, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic bowel diseases, inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal bums, coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, ulcerative colitis, migraine, rhinitis, eczema, interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome, diabetic nephropathy, multiple myositis, Guillain-Baπe syndrome, Meniere's disease, polyneuritis, multiple neuritis, mononeuritis, radiculopathy, hyperthyroidism, Basedow's disease, pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia, anerythroplasia, osteoporosis, sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photoallergic sensitivity, cutaneous T cell lymphoma, arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, myocardosis, scleroderma, Wegener's granuloma, Sjogren's syndrome, adiposis, eosinophilic fascitis, lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis, glomeralonephritis, male pattern alopecia or alopecia senilis by preventing epilation or providing hair germination and/or promoting hair generation and hair growth, muscular dystrophy, pyoderma and Sezary's syndrome, Addison's disease, ischemia-reperfusion injury of organs which occurs upon preservation, transplantation or ischemic disease, endotoxin-shock, pseudomembranous colitis, colitis caused by drag or radiation, ischemic acute renal insufficiency, chronic renal insufficiency, toxinosis caused by lung-oxygen or drags, lung cancer, pulmonary emphysema, cataracta, siderosis, retinitis pigmentosa, senile macular degeneration, vitreal scarring, comeal alkali bum, dermatitis eiythema multiforme, linear IgA ballous dermatitis and cement dermatitis, gingivitis, periodontitis, sepsis, pancreatitis, diseases caused by environmental pollution, aging, carcinogenesis, metastasis of carcinoma and hypobaropathy, disease caused by histamine or leukotriene-C4 release, Behcet's disease, autoimmune hepatitis, primary biliary cinhosis, sclerosing cholangitis, partial liver resection, acute liver necrosis, necrosis caused by toxin, viral hepatitis, shock, or anoxia, B-virus hepatitis, non-
A/non-B hepatitis, cinhosis, alcoholic cinhosis, hepatic failure, fulminant hepatic failure, late-onset hepatic failure, "acute-on-chronic" liver failure, augmentation of chemotherapeutic effect, cytomegalovirus infection, HCMV infection, AIDS, cancer, senile dementia, trauma, and chronic bacterial infection. The compounds of the present invention are also useful for treating or preventing Alzheimer's Disease.
Also embodied within the present invention is a method of preventing or treating resistance to transplantation or transplantation rejection of organs or tissues in a mammalian patient in need thereof, which comprises administering a therapeutically effective amount of the compound of Formula I.
A method of suppressing the immune system in a mammalian patient in need thereof, which comprises administering to the patient an immune system suppressing amount of the compound of Formula I is yet another embodiment.
Most particularly, the method described herein encompasses a method of treating or preventing bone manow or organ transplant rejection which is comprised of admininstering to a mammalian patient in need of such treatment or prevention a compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof, in an amount that is effective for treating or preventing bone manow or organ transplant rejection.
The compounds of the present invention are also useful for treating a respiratory dieases or condition, such as asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia
Furthermore, the compounds of the present invention are selective agonists of the SlPi/Edgl receptor having selectivity over SlP3/Edg3 receptor. An Edgl selective agonist has advantages over cuπent therapies and extends the therapeutic window of lymphocytes sequestration agents, allowing better tolerability with higher dosing and thus improving efficacy as monotherapy.
The present invention also includes a pharmaceutical formulation comprising a pharmaceutically acceptable carrier and the compound of Formula I or a pharmaceutically acceptable salt or hydrate thereof. A prefeπed embodiment of the formulation is one where a second immunosuppressive agent is also included. Examples of such second immunosuppressive agents are, but are not limited to azathioprine, brequinar sodium, deoxyspergualin, mizaribine, mycophenolic acid moφholino ester, cyclosporin, FK-506, rapamycin, FTY720 and ISAtx247 (Isotechnika). Methods of co-administering a compound of Formula I with a second immunosuppressive agent, including one or more of the above, is also encompassed within the invention. The present compounds, including salts and hydrates thereof, are useful in the treatment of autoimmune diseases, including the prevention of rejection of bone manow transplant, foreign organ transplants and/or related afflictions, diseases and illnesses. The compounds of this invention can be administered by any means that effects contact of the active ingredient compound with the site of action in the body of a warm-blooded animal. For example, administration can be oral, topical, including tiansdermal, ocular, buccal, intianasal, inhalation, intravaginal, rectal, intracisternal and parenteral. The term "parenteral" as used herein refers to modes of administration which include subcutaneous, intravenous, intramuscular, intraarticular injection or infusion, intrasternal and intraperitoneal.
The compounds can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but are generally administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
The dosage administered will be dependent on the age, health and weight of the recipient, the extent of disease, kind of concuπent treatment, if any, frequency of tieatment and the nature of the effect desired. Usually, a daily dosage of active ingredient compound will be from about 0.1 -2000 milligrams per day.
Ordinarily, from 1 to 100 milligrams per day in one or more applications is effective to obtain desired results. These dosages are the effective amounts for the treatment of autoimmune diseases, the prevention of rejection of foreign organ transplants and/or related afflictions, diseases and illnesses. The active ingredient can be administered orally in solid dosage forms, such as capsules, tablets, troches, dragees, granules and powders, or in liquid dosage forms, such as elixirs, syrups, emulsions, dispersions, and suspensions. The active ingredient can also be administered parenterally, in sterile liquid dosage forms, such as dispersions, suspensions or solutions. Other dosages forms that can also be used to administer the active ingredient as an ointment, cream, drops, tiansdermal patch or powder for topical administration, as an ophthalmic solution or suspension formation, i.e., eye drops, for ocular administration, as an aerosol spray or powder composition for inhalation or intranasal administration, or as a cream, ointment, spray or suppository for rectal or vaginal administration.
Gelatin capsules contain the active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar diluents can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar coated or film coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration can contain coloring and flavoring to increase patient acceptance.
In general, water, a suitable oil, saline, aqueous dextrose (glucose), and related sugar solutions and glycols such as propylene glycol or polyethylene gycols are suitable carriers for parenteral solutions. Solutions for parenteral administration preferably contain a water soluble salt of the active ingredient, suitable stabilizing agents, and if necessary, buffer substances. Antioxidizing agents such as sodium bisulfite, sodium sulfite, or ascorbic acid, either alone or combined, are suitable stabilizing agents. Also used are citric acid and its salts and sodium EDTA. In addition, parenteral solutions can contain preservatives, such as benzalkonium chloride, methyl- or propylparaben, and chlorobutanol.
Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, A. Osol, a standard reference text in this field.
For administration by inhalation, the compounds of the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers. The compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device. The prefened delivery system for inhalation is a metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons.
For ocular administration, an ophthalmic preparation may be formulated with an appropriate weight percent solution or suspension of the compounds of Formula I in an appropriate ophthalmic vehicle, such that the compound is maintained in contact with the ocular surface for a sufficient time period to allow the compound to penetrate the comeal and internal regions of the eye.
Useful pharmaceutical dosage-forms for administration of the compounds of this invention can be illustrated as follows: CAPSULES
A large number of unit capsules are prepared by filling standard two- piece hard gelatin capsules each with 100 milligrams of powdered active ingredient, 150 milligrams of lactose, 50 milligrams of cellulose, and 6 milligrams magnesium stearate. SOFT GELATIN CAPSULES
A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive displacement pump into gelatin to form soft gelatin capsules containing 100 milligrams of the active ingredient. The capsules are washed and dried.
TABLETS A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 milligrams of active ingredient, 0.2 milligrams of colloidal silicon dioxide, 5 milligrams of magnesium stearate, 275 milligrams of microcrystalline cellulose, 11 milligrams of starch and 98.8 milligrams of lactose. Appropriate coatings may be applied to increase palatability or delay absorption.
INJECTABLE A parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol. The solution is made to volume with water for injection and sterilized.
SUSPENSION An aqueous suspension is prepared for oral administration so that each 5 milliliters contain 100 milligrams of finely divided active ingredient, 100 milligrams of sodium carboxymethyl cellulose, 5 milligrams of sodium benzoate, 1.0 grams of sorbitol solution, U.S.P., and 0.025 milliliters of vanillin. The same dosage forms can generally be used when the compounds of this invention are administered stepwise or in conjunction with another therapeutic agent. When drugs are administered in physical combination, the dosage form and administration route should be selected depending on the compatibility of the combined drugs. Thus the term coadministration is understood to include the administration of the two agents concomitantly or sequentially, or alternatively as a fixed dose combination of the two active components.
METHODS OF SYNTHESIS
Methods for preparing the compounds of this invention are illustrated in the following examples. Alternative routes will be easily discernible to practitioners in the field.
A convenient method to prepare the compounds of the general structure i in the present invention is shown in Scheme 1. Carboxylic acid ii can be activated for acylation with a reagent such as N,N'-dicyclohexylcarbodiimide, l-(3- dimethylaminopropyl)-3-ethylcarbodiimide, 1,1 '-carbonyldiimidazole, or bis(2-oxo-3- oxazolidinyl)phosphinic chloride in the presence of a suitable base (if necessary) such as triethylamine, N,N-diisopropylethylamine, or sodium bicarbonate in a solvent such as 1,2-dichloroethane, toluene, xylenes, N,N-dimethylformamide or N-methyl pynolidinone. A 2-(amino)aryl N-hydroxyamidine of general structure iii can then be added which results in the formation of an acyl N-hydroxyamidine iv. This intermediate can be isolated using methods known to those skilled in the art (e.g., crystallization, silica gel chromatography, HPLC) and in a subsequent step, cyclized/dehydrated by warming iv in a suitable solvent (e.g., 1,2-dichloroethane, toluene, xylenes, N,N-dimethylformamide or N-methyl pynolidinone) to give a 1,2,4- oxadiazole of structure i. Conversion of iii to iv may require added base, in which case reagents such as pyridine, N,N-diisopropylethylarnine or tetrabutylammonium fluoride can be used. It may be more convenient or desirable to not isolate N- hydroxyamidine iv, in which case the teansformation of ii to i can be carried out as a continuous process.
It possible to use acylating agents other than activated carboxylic acid ii to give compounds i. Specifically, it might be advantageous or desirable to use a carboxylic acid chloride, carboxylic acid anhydride, carboxamide or carbonitrile in the place of carboxylic acid ii and an acyl activating agent to prepare 1,2,4-oxadiazole compounds i as described above. Methods to prepare 1,2,4-oxadiazoles using these other acylating agents as well as other methods pertinent to the present invention are known to those skilled in the art and have been reviewed in the literature (see, Clapp, L.B., "1,2,3- and 1,2,4-Oxadiazoles", pp. 366-91 in Comprehensive Heterocyclic Chemistry, Volume 6, Potts, K. T., Editor, Pergamon Press, 1984). Scheme 1
Figure imgf000048_0001
solvent, base
solvent, base, heat
Figure imgf000048_0002
X, Y, Z = N or C-R8, A = N or CR°, D = N or C-R* E= N or C-R6 and G = N or C-R7
A second method that can be used to prepare the compounds of the general stracture i in the present invention is shown in Scheme 2. Carboxylic acid ii is activated as described in Scheme 1 and used to acylate a 2-(substituted)aryl N- hydroxyamidine v in which the functional group X is a leaving group such as fluoro, chloro, bromo, iodo, cyano, alkylsulfonyloxy or arylsulfonyloxy. Conversion to compound iv and ring closure to give i is effected using the methods described above. Displacement of the leaving group X is carried out by treating vi with ammonia, an alkylamine or a dialkylamine in a suitable solvent (e.g., methanol, ethanol, N,N- dimethylformamide, dimethylsulfoxide) at or above ambient temperature to give 1,2,4-oxadiazole i. Alternatively, as reported by Park and Cho in Tetrahedron Letters, 1997, 38, 8331-34, vi can be treated with N-methylformamide in the presence of diethanolamine at elevated temperature to give compounds i, where -NR1R2 is - NHCH3.
Scheme 2
R
3) solvent, heat
Figure imgf000049_0002
-met y ormam e diethanolamine heat X, Y, Z = N or C-R8, A = N or CR3, D = N or C-R'
(R1 = H, R2 = -CH3) E= N or C-R6 and G = N or C-R7
It is understood that the chemical stracture(s) of groups R3-R8 can be manipulated in compounds i. Examples of this include (but are not limited to): 1) if one or more of R3-R8 is -OH, treatment of i with an alkyl halide or alkyl sulfonate ester in the presence of an appropriate base (e.g., N,N-diisopropylethylamine, triethylamine, pyridine, sodium carbonate) in a suitable solvent (methylene chloride, acetonitrile, toluene, N,N-dimethylformamide) at or above ambient temperature can give compounds i in which one or more of R3-R8 is alkoxy; 2) if one or more of R3- R8 is -CI, -Br, -I, or -OSO2CF3, treatment of i with an aryl boronic acid and a suitable base (sodium hydroxide, potassium bicarbonate) in the presence of a palladium catalyst (e.g., tetrakis(triphenylphosphine) palladium or dichloropalladium bis(triphenylphosphine) in a suitable solvent (e.g., ethanol, N,N-dimethylformamide, dioxane, toluene) at or above ambient temperature can give compounds i in which one or more of R3-R8 is aryl.
A convenient method to prepare the N-hydroxyamidine intermediates iii or v used to prepare the compounds of the present invention is shown in Scheme 3. For either intermediate, the coπesponding carbonitrile viii or ix is treated with hydroxylamine (from aqueous hydroxylamine solution or generated by treating hydroxylamine hydrochloride with a base such as triethylamine, N,N- diisopropylethylamine, or sodium bicarbonate) in an appropriate solvent (methanol, ethanol, water, N,N-dimethylformamide) at or above ambient temperature. Many of the carbonitriles viii or ix as well as carboxylic acids ii are available from commercial sources or can be prepared by those skilled in the art. using reported literature procedures.
Scheme 3
NHpOH, solvent
Figure imgf000050_0001
Figure imgf000050_0002
viii iii
NH2OH, solvent
Figure imgf000050_0003
Figure imgf000050_0004
Q = -F, -CI, -Br, -I, -OS02R Q = -F, -CI, -Br, -I, -OS02R
Figure imgf000050_0005
While the general structure i is achiral, it is understood that any of groups R1-R8 may have asymmetric centers, in which case the individual stereoisomers of i can obtained by methods known to those skilled in the art which include (but are not limited to): stereospecific synthesis, resolution of salts of i or any of the intermediates used in its preparation with enantiopure acids or bases, resolution of i or any of the intermediates used in its preparation by HPLC employing enantiopure stationary phases.
REPRESENTATIVE EXAMPLES
Compounds of the invention are exemplified as follows:
GENERAL Concentration of solutions was carried out on a rotary evaporator under reduced pressure. Conventional flash chromatography was carried out on silica gel (230-400 mesh). Flash chromatography was also carried out using a Biotage Flash Chromatography apparatus (Dyax Corp.) on silica gel (32-63 mM, 60 A pore size) in pre-packed cartridges of the size noted. NMR spectra were obtained in CDCI3 solution unless otherwise noted. Coupling constants (J) are in hertz (Hz).
Abbreviations: diethyl ether (ether), triethylamine (TEA), N,N-diisopropylethylamine (DIEA) sat'd aqueous (sat'd), it (rt), hour(s) (h), minute(s) (min).
HPLC Methods
HPLC A: YMC ODS A, 5μ, 4.6 x 50 mm column, gradient 10:90-95:5 v/v CH3CN:H2O + 0.05% TFA over 4.5 min, then hold at 95:5 v/v CH3CN:H2O +
0.05% TFA for 1.5 min; 2.5 mL/min, diode aπay detection 200-400 nM
HPLC B: Analytical Sales & Service ARMOR C 18 5 m 2 x 25 cm column, gradient 10:90-100:0 v/v CH3CN:H2θ + 0.05% TFA over 15 min, then hold at 100.0 v/v CH3CN:H2θ + 0.05% TFA for 10 min; 20 mL/min, diode anay detection 200-400 nM
PREPARATION OF N-HYDROXYAMIDINE INTERMEDIATES
N-HYDROXYAMIDINE 1 2-Chloro-N-hvdroxy-nicotinamidine A mixture of 2-chloro-3-pyridine-carbonitrile (5.00 g, 37 mmol), hydroxylamine hydrochloride (3.73 g, 54 mmol) and sodium bicarbonate (9.10 g, 108 mmol) were stiπed together in CH3OH (250 ml) at 50°C for 16 h. The reaction was cooled, filtered, washed with CH2CI2 and the filtrate concentrated to give a yellow solid: lH NMR (500 MHz , CD3OD) δ 8.43 (dd, J=1.9, 7.6 ,1H), 7.88 (dd, J=1.9, 7.6 ,1H), 7.44 (dd, J=4.5, 7.5 ,1H).
N-HYDROXYAMIDINEs 2-6 The following N-HYDROXYAMIDINE intermediates were prepared using a procedure analogous to that described for N-HYDROXYAMIDINE 1 substituting the appropriate nitrile for 2-chloro-3-pyridine-carbonitrile.
Figure imgf000052_0001
Figure imgf000053_0001
N-HYDROXYAMIDINE 7 2-(N-Methylamino)-N-hydroxy-nicotinamidine A mixture of 10 g (72 mmol) 2-chloro-3-pyridine-carbonitrile, 40 mL of 40% methylamine in H2O and 20 mL of iPrOH was stiπed at 55 °C for 1.5 h. Aqueous hydroxylamine (6.0 mL, 50 wt. % in H2O) was added and the resulting mixture was stiπed at 55 °C for 1 h. The solution was cooled to rt. The solid that precipitated was filtered, washed with 50 mL of cold (0 °C) 1 : 1 v/v iPrOH/ H2O and dried to afford 7.52 g of the title compound: iH NMR (500 MHz , DMSO) δ 9.88 (br s, IH), 8.11 (q, J= 4.5, IH), 8.02 (dd, J= 1.5, 5.0, IH), 7.75 (dd, J = 1.5, 7.5, IH), 6.54 (dd, J = 5.0, 7.5, IH), 5.90 (s, 2H), 2.89 (d, J= 4.5, 3H); ESI-MS 167 (M+H).
N-HYDROXYAMIDINE 8 2-(Amino)-N-hydroxy-nicotinamidine
A solution of 2-amino-3-pyridine-carbonitrile (0.50 g, 4.2 mmol) and hydroxylamine (0.42 g, 50% in H2O) in 10 mL of MeOH was stiπed at 50°C for 16 h. The reaction was cooled and concentrated. Chromatography on a Biotage 40S cartridge using EtOAc as the eluant afforded 0.50 g of the title compound: iH NMR (500 MHz , CD3OD) δ 7.91 (dd, J=1.8, 5.0, IH), 7.76 (dd, J=1.9, 7.8, IH), 6.66 (dd,
J=5.2, 7.7, IH). N-HYDROXYAMIDINE 9 3-(N-Methylamino)-pyrazine-2-(N-hydroxyamidine) Step A: 2-(N-Methylamino)-3-cyanopyrazine
A solution of 3.0 mL of 40% aqueous methyl amine and 3.8 mL (27 mmol) of TEA in 20 mL of THF was added dropwise to a solution of l.35 g (10.4 mmol) of pyrazine-2,3-dicarbonitrile in 25 mL of THF over 45 min. The resulting mixture was stiπed for 15 min, then concentrated. The residue was partitioned between 100 mL of CH2CI2 and 50 mL of 1 N HC1. The layers were separated and the aqueous layer was extracted with 100 mL of CH2CI2. The organic extracts were combined, dried and concentrated. Chromatography on a Biotage 40S cartridge using 9: 1 v/v hexanes/EtOAc as the eluant afforded 446 mg of the title compound: ESI-MS 135 (M+H); HPLC A: 3.03 min.
Step B: 3-(N-Methylamino)-pyrazine-2-(N-hydroxyamidine) A mixture of 446 mg (3.3 mmol) of 3 -(N-methylamino)-pyrazfne-2-(N- hydroxyamidine) (from Step A), 486 mg (7 mmol) of hydroxylamine hydrochlori.de and 1.2 mL (7 mmol) of DIEA in 15 mL of EtOH was heated at reflux for 30 min. The mixture was cooled to 0 °C. The solid that precipitated was filtered, rinsed with cold EtOH and dried to afford 340 mg of the title compound: lH NMR (500 MHz , DMSO) δ 10.2 (s, IH), 8.36 (q, J= 4.5, IH), 8.07 (d, J= 2.5, IH), 7.77 (d, J= 2.5, IH), 5.97 (s, 2H), 2.93 (d, J = 4.5, 3H); ESI-MS 135 (M+H).
N-HYDROXYAMIDINE 10 2-(N-Methylamino)-5-fluoro-N-hvdroxynicotinamidine Step A: 2,6-Dichloro-5-fluoronicotinamide
To a mixture of 2,6-dichloro-5-fluoronicotinamide (5.50 g, 26.2 mmol) in dichloromethane (50 mL) and dimethylformamide (2 drops) cooled to 0 °C, oxalyl chloride (6.72 mL, 78.6 mmol) was added dropwise, and the cooling bath was removed. After 2 hr, the reaction mixture was concentrated in vacuo, and the residue azeotioped with toluene (1 X 10 mL). The resultant brown residue was dissolved in dioxane (50 mL) and concentrated NH OH was added dropwise. The mixture was stiπed at ambient temperature for 16 h, concentrated in vacuo and triturated from 50%Et2O/z-PrOH (30 mL) at 0 °C to give 5.48 g of the title compound as a beige solid: !H NMR (500 MHz , CDC13) δ 6.27 (br, 1 H), 6.78 (br, 1 H), 8.11 (d, IH, J= 7.3 Hz).
Step B: 2-Chloro-5-fluoronicotinamide Under a N2 atmosphere, 2,6-dichloro-5-fluoronicotinamide (500 mg,
2.39 mmol), potassium acetate (258 mg, 2.63 mmol), and PtO2 (25 mg) were combined. EtOAc (2.5 mL) and CH3OH (2.5 mL) were then added, followed by one atmosphere of hydrogen via balloon. After 26 hr, the reaction mixture was filtered through Celite®, and concentrated in vacuo. The residue was treated with EtOAc (10 mL), filtered, and the filtrate concentrated in vacuo. Purification of the residue by flash chromatography (1, 2% CH3OH/CH2Cl2) on SiO2 afforded 130 mg of the title compound as a white solid: 1H NMR (500 MHz , CD3OD) δ 7.79 (dd, IH, J= 2.8, 7.7 Hz), 8.37 (d, lH, J= 2.8 Hz).
Step C: 2-Chloro-5-fluoropyridine-3-carbonitrile
To a mixture of 2-chloro-5-fluoronicotinamide (880 mg, 5.04 mmol), triethylamine (1.55 mL, 11.1 mmol) and dichloromethane (15 mL) cooled to 0 °C, trifluoroacetic anhydride (783 μL, 5.55 mmol) was added dropwise. The resultant yellow solution was stiπed for 1 hr at 0 °C, diluted with dichloromethane (5 mL) and washed with saturated NaHCO3 (2 X 10 mL), brine (1 X 10 mL) and dried over MgSO4. The mixture was filtered, concentrated in vacuo, and purified by flash chromatography (5, 10% EtOAc/hexanes) on SiO2 to afford 770 mg of the title compound as a white solid: 1H NMR (500 MHz , CDC13) δ 7.77 (dd, 1 H J= 3.0, 6.9 Hz), 8.49 (d, 1 H/= 3.0 Hz); 13C NMR (500 MHz, CDC13) δ 111.3, 113.3, 129.4 (J = 21.1 Hz), 141.5, (J= 26.9 Hz), 147.6, 157.2 (J= 260 Hz).
Step D: 5-Fluoro-2-methylaminopyridine-3-carbonitrile
In a sealed tube, 2-chloro-5-fluoropyridine-3-carbonitrile (59 mg, 0.377 mmol) was dissolved in dioxane (1.5 mL). A 2.0 M solution of methylamine in THF (283 μL, 0.565 mmol) was added, the tube was sealed and heated to 60 °C. After 3 hr, additional methylamine in THF (283 μL, 0.565 mmol) was added, and the reaction mixture was heated for 16 hr. The reaction mixture was cooled to ambient temperature, concentrated in vacuo, and purified by flash chromatography (5, 7, 10% EtOAc/hexanes) on SiO2 to afford 21 mg of the title compound as a white film: !H NMR (500 MHz , CD3OD) δ 2.93, (s, 3 H), 7.68 (dd, 1 HJ= 3.0, 7.9 Hz), 8.19 (d, 1 H J= 3.0, Hz); HPLC/MS (HPLC A): 152 (M+H)+, 1.97 min.
Step E: 2-(N-Methylamino)-5-fluoro-N-hydroxynicotinamidine
To a solution of 5-fluoro-2-methylaminopyridine-3-carbonitrile in absolute ethanol (1 mL) and triethylamine (28 μL, 0.198 mmol), hydroxylamine hydrochloride (12 mg, 0.172 mmol) was added and the mixture was heated to reflux. After 6 hr, the reaction mixture was cooled to ambient temperature, concentrated in vacuo and purified by flash chromatography (10, 20, 40 % EtOAc/hexanes) on SiO2 to afford 8.0 mg of the title compound as a white film: HPLC/MS (HPLC A): 152 (M+H)+, 0.33 min.
N-HYDROXYAMIDINE 11
2-Amino-5-fluoro-N-hvdroxynicotinamidine Step A: 2-Amino-5-fluoropyridine-3-carbonitrile
In a sealed tube, concentrated ammonia (0.6 mL) was added to a solution of 2-chloro-5-fluoropyridine-3-carbonitrile (100 mg, 0.639 mmol, from Ν- HYDROXYAMIDINE 10, Step C) in dioxane (1 mL) and the reaction mixture was heated to 110 °C. After 5 hr, the reaction mixture was cooled to ambient temperature, concentrated in vacuo and the residue purified by flash chromatography (10, 20, 30, 50% EtOAc/hexanes) to afford 31 mg of the title compound was a white film (35%): *H NMR (500 MHz , CDC13) δ 5.16 (br, 2 H), 7.45 (dd, 1 H, J= 2.4, 7.6 Hz), 8.15 (d, l H, J= 2.1 Hz).
Step B: 2-Amino-5-fluoro-N-hydroxynicotinamidine
To a solution of 2-amino-5-fluoropyridine-3-carbonitrile (38 mg, 0.277 mmol) in ethanol (2 mL) and triethylamine (58 μL, 0.416 mmol), hydroxylamine hydrochloride (23 mg, 0.333 mmol) was added and the mixture was heated to reflux.
After 6 hr, the reaction mixture was cooled to ambient temperature, concentrated in vacuo and purified by flash chromatography (30, 50 % EtOAc/hexanes) on SiO2 to afford 35 mg of the title compound as a white film (74%): HPLC/MS (HPLC A): 171 (M+H)+.
PREPARATION OF CARBOXYLIC ACID INTERMEDIATES
CARBOXYLIC ACID 1 3 -Fluoro-4-cvclopentyl-benzoic acid A solution of 0.45 g (1.45 mmol) of benzyl 3-fluoro-4-bromo-benzoate (0.45 g, 1.45 mmol) in 4.4 mL of 0.5 M cyclopentylzinc bromide solution in THF) was treated with ~5 mg of bis(tri-t-butylphosphine)palladium(0) and the resulting mixture was stined at rt for 24 h. The reaction mixture was directly purified on a Biotage 40S cartridge using 1 : 1 hexanes/EtOAc as the eluant. A mixture of the resulting solid (0.27 g, 0.91 mmol) and 10%> Pd/C in 5 mL of MeOH was stined under 1 atin of H2 for 3 h. The reaction was filtered and concentrated. Purification by HPLC B afforded the title compound: Η NMR (500 MHz , CDCI3) δ 7.83 (dd, J=1.6, 8.0, IH), 7.72 (dd, J=1.6, 10.5, IH), 7.36 (t, J=7.7, IH), 3.30 (m, IH), 2.05- 2.14 (m, 2H), 1.58-1.90 (m, 6H).
CARBOXYLIC ACID 2
(+/-)-4-( 1 -Oxo-2-methylbutyl)benzoic acid Step A: (+/-)-Ethyl 4-(l-oxo-2-methylbutyl)benzoate
A solution of 0.58 g (4.5 mmol) of (+/-)-2-methylbutyryl chloride in 10 mL of 0.5 M 4-(ethoxycarbonyl)phenylzinc iodide solution in THF) was treated with ~5 mg of bis(tri-t-butylphosphine)palladium(0) and the resulting mixture was stined at rt for 1 h. The reaction mixture was partitioned between 50 mL of EtOAc ethyl acetate and 25 mL of 2 N HC1 and the layers were separated. The organic layer was washed with 25 mL of sat'd NaCl, dried and concentrated. Silica gel chromatography using 15:1 v/v hexanes/ethyl acetate (15:1) as the eluant afforded the title compound: Η NMR (500 MHz , CDCI3) δ 8.12 (d, J= 8.4, 2H), 7.98 (d, J= 8.5, 2H), 4.40 (q, J=
7.2, 2H), 3.40 (m, IH), 1.83 (m, IH), 1.51 (m, IH), 1.41 (t, J= 7.2, 3H), 1.20 (d, J= 6.8 3H), 0.91 (t, J= 7.5 3H). Step B: (+/-)-4-(l-Oxo-2-methylbutyl)benzoic acid
A solution of 0.57 g (2.4 mmol) of (+/-)-ethyl 4-(l-oxo-2- methylbutyl)benzoate (from Step A) in 10 mL of MeOH, 3 mL of THF and 2.4 mL of 5 N NaOH was stined at rt for 16 h. The mixture was diluted with 20 mL of H2O and extracted with 25 mL of CH2CI2. The aqueous layer was acidified (pH 1) and extracted with 50 mL of EtOAc. The organic layer was washed with 25 mL of sat'd NaCl, dried and concentrated to give 0.41 g of the title compound: Η NMR (500 MHz , CDCI3) δ 8.21 (d, J= 8.4, 2H), 8.03 (d, J= 8.5, 2H), 3.41 (m, IH), 1.85 (m,
IH), 1.52 (m, IH), 1.21 (d, J= 6.9, 3H), 0.93 (t, J= 7.5, 3H).
CARBOXYLIC ACID 3 4-(l -Oxo-2-methylpropyl)benzoic acid The title compound was prepared using procedure analogous to that described for CARBOXYLIC ACID 2 substituting isobutyryl chloride for (+/-)-2- methylbutyryl chloride in Step A: Η NMR (500 MHz, CDCI3) δ 8.21 (d, J= 8.5,
2H), 8.03 (d, J= 8.5, 2H), 3.57 (m, IH), 1.24 (d, J= 6.9, 6H).
CARBOXYLIC ACID 4 4-(Cyclobutyldifluoromethyl)benzoic acid Step A: Ethyl 4-(cyclobutylcarbonyl)benzoate
The title compound was prepared using procedure analogous to that described for CARBOXYLIC ACID 2, substituting cyclobutanecarbonyl chloride for (+/-)-2-methylbutyryl chloride in Step A: 'H NMR (500 MHz , CDCI3) δ 8.10 (d, J=
8.2 , 2H), 7.93 (d, J= 8.5 , 2H), 4.40 (q, J= 7.2 , 2H), 4.01 (m, IH), 2.37-2.46 (m, 2H), 2.28-2.36 (m, 2H), 2.04-2.15 (m, IH), 1.88-1.97 (m, IH), 1.41 (t, J= 7.1 , 3H).
Step B: Ethyl 4-(cyclobutyldifluoromethyl)benzoate
A solution of 810 mg (3.5 mmol) of ethyl 4- (cyclobutylcarbonyl)benzoic acid (from Step A) in 5 mL of toluene was treated with 1.30 g (5.9 mmol) of [bis(2-methoxyethyl)amino]sulfur trifluoride and 0.41 mL (0.7 mmol) of EtOH and the resulting mixture was heated to 80°C for 18 h. The reaction was concentrated. Silica gel chromatography using 20: 1 v/v hexanes/EtOAc afforded the title compound: Η NMR (500 MHz , CDCI3) δ 8.07 (d, J= 8.2 , 2H), 7.51 (d, J= 8.5 , 2H), 4.39 (q, J= 7.2 , 2H), 2.96 (m, IH), 2.15-2.27 (m, 2H), 1.80-1.99 (m, 4H), 1.40 (t, J= 7.1 , 3H).
Step C: 4-(Cyclobutyldifluoromethyl)benzoic acid A solution of360 mg (1.4 mmol) of ethyl 4-
(cyclobutyldifluoromethyl)benzoate (from Step B) in 4 mL of 1 : 1 v/v MeOH/THF was treated with 2.1 mL of 1.0 N NaOH. The resulting mixture was stiπed at 50°C for 3 h at, then cooled and concentrated. The residue was partitioned between EtOAc and 2 N HC1. The organic layer was washed with 2 N HC1 (25 ml), 25 mL of sat'd NaCl, dried and concentrated to give 280 mg of the title compound: Η NMR (500 MHz , CDCI3) δ 8.15 (d, J= 8.5 , 2H), 7.56 (d, J= 8.4 , 2H), 2.97 (m, IH), 2.17-2.27
(m, 2H), 1.80-2.02 (m, 4H).
CARBOXYLIC ACID 5 4-(l , 1 -Difluoro-2-methylpropyl)benzoic acid
The title compound was prepared using procedure analogous to that described for CARBOXYLIC ACID 4 substituting ethyl 4-
(isopropylcarbonyl)benzoate for ethyl 4-(cyclobutylcarbonyl)benzoate in Step B: Η NMR (500 MHz , CDCI3) δ 8.17 (d, J= 8.3 , 2H), 7.56 (d, J= 8.4 , 2H), 2.34 (m, IH), 1.00 (d, J= 6.8 , 6H).
CARBOXYLIC ACID 6 3 -Fluoro-4-(2-methylpropionyl)benzoic acid Step A: l-Bromo-3-fluoro-4-(2'-methyl)propiophenone A solution of 1.00 g (3.8 mmol) of N-methoxy-N-methyl (4-bromo-2- fluoro)benzamide in 10 mL of THF at -78 °C was treated with 2.3 mL of 2.0 M isopropylmagnesium chloride solution in THF. The reaction was allowed to warm to rt and was stiπed for 3 h. The reaction was diluted with 50 mL of ethyl ether, washed with 25 mL of 2 N HC1, 25 mL of sat'd NaCl, dried and concentrated. Silica gel chromatography using 50: 1 hexanes/EtOAc as the eluant gave 143 mg of the title compound: Η NMR (500 MHz , CDCI3) δ 7.67 (t, J= 8.2 , IH), 7.38 (dd, J= 1.8, 8.4
, IH), 7.33 (dd, J= 1.6, 10.3, IH), 3.35 (m, IH), 1.19 (d, J= 6.9 , 6H). Step B: 3-Fluoro-4-isobutyrylbenzoic acid
A solution of 143 mg (0.58 mmol) of l-bromo-3-fluoro-4-(2' -methyl) propiophenone (from Step A), 41 mg (0.35 mmol) of zinc cyanide, 11 mg (0.011 mmol) of tris(dibenzylideneacetone)-dipalladium(0) and 15 mg (0.026 mmol) of 1,1- bis(diphenylphosphino)-feπocene (15 mg, 0.026 mmol) in 2 mL of DMF and 0.030 mL water was heated to 85°C for 3 h. The reaction was cooled, loaded onto silica gel and eluted with hexane/ethyl acetate (20: 1) to give the product as a yellow solid (36 mg). A solution of this solid in methanol (2 L) was treated with excess 5 N NaOH and heated at 60°C for 3 h. The reaction was cooled, diluted with 50 mL of EtOAc, washed with 25 mL of 2 N HCl, dried and concentrated to give the title compound.
CARBOXYLIC ACID 7 3 -Trifluoromethyl-4-(2-(S)-butoxy)benzoic acid Step A: 3-Trifluoromethyl-4-(2-(S)-butoxy)benzonitrile A solution of 1.1 g (5.9 mmol) of 4-fluoro-3 - trifluoromethylbenzonitrile and 485 mg (6.5 mmol) of (S)-(+)-2-butanol in 10 mL of THF at-10°C was treated with 235 mg (5.9 mmol) of sodium hydride. The resulting mixture was stined cold for 2 h, then quenched with 10 mL of H2O. The quenched solution was extracted with 30 mL of Et2θ, dried over MgSO4 and concentrated. Chromatography on a Biotage 40M cartridge using 4: 1 v/v hexanes/Ethyl acetate as the eluant afforded 550 mg of the title compound: iH NMR (500 MHz) δ 0.99 (t, J= 7.6, 3H), 1.35 (d, J= 6.2, 3H), 1.58-1.83 (m, 2H), 4.51 (septet, IH), 7.04 (d, J= 8.7, IH), 7.75 (d, J= 8.7, IH), 7.85 (s, IH).
Step B: 3-Trifluoromethyl-4-(2-(S)-butoxy)benzoic acid
A solution of 550 mg (2.2 mmol) of 3-trifluoromethyl-4-(2-(S)- methylpropyloxy) benzonitrile (from Step A) in 5 mL of ethanol was treated with 1.5 mL of 5.0 N NaOH and was heated to 80°C for 3 h. The reaction was then concentrated, treated with 2 N HCl, extracted with 30mL of EtOAc, dried and concentrated to afford 600 mg of the title compound: lH NMR (500 Mhz) δ 0.99 (t, J= 7.3, 3H), 1.43 (d, J= 5.9, 3H), 1.73-1.83 (m, 2H), 4.54 (septet, IH), 7.02 (d, J= 8.9, IH), 8.21 (d, J= 8.9, IH), 8.32 (s, IH). CARBOXYLIC ACIDs 8-14 The following intermediates were prepared using procedures analogs to those described for CARBOXYLIC ACID 7 substituting the appropriate alcohol for (S)-2- butanol in Step A.
Figure imgf000061_0001
Figure imgf000061_0002
Figure imgf000062_0001
CARBOXYLIC ACID 15 3-Trifluoromethyl-4-(l-(S)-met3 yl-2.2.2-trifluoroethoxy)benzoic acid Step A: l-(S)-Methyl-2,2,2-trifluoroethanol The title compound was prepared using the procedure reported by
Ramachandran, P. V., et.al. in Tetrahedron, 1993, 49(9), 1725-38.
Step B: 3-Trifluoromethyl-4-(l-(S)-methyl-2,2,2-trifluoroethoxy)benzoic acid
The title compound was prepared using procedures analogous to those described for CARBOXYLIC ACID 7 substituting 1 -(S)-mefhyl-2,2,2-trifluoroethanol (from Step A) for (S)-2-butanol in CARBOXYLIC ACID 7, Step A. The enantiomeric purity of the title compound was determined by converting it to the conesponding methyl ester (excess 2.0 M trimethylsilyldiazomethane solution in cyclohexane, THF/MeOH, 5 min) and assaying by HPLC. Conditions: Chiralcel OD 4.6 x 250 mm column, 98:2 v/v heptane/iPrOH, 1.0 mL/min, λ - 254 nM. (R)- enantiomer = 8.5 min, (S)-enantiomer = 10.4 min.
CARBOXYLIC ACID 16 3 -Fluoro-4-(2-(S)-butoxy)benzoic acid Step A: 3-Fluoro-4-(2-(S)-butoxy)benzaldehyde
A solution of 750 mg (5.4 mmol) of 3-fluoro-4-hydroxybenzaldehyde, 403 mg (5.4 mmol) of (R)-(-)-2-butanol and 2 g (7.5 mmol) triphenylphosphme in 10 mL of THF was treated with 1.5 mL of diisopropylazodicarboxylate. The resulting Step C: 3,5-Difluoro-4-(2-(S)-butoxy)benzoic acid
The title compound was prepared using procedure analogous to that described in CARBOXYLIC ACID 7, Step B substituting 3,5-difluoro-4-(2-(S)- butoxy)benzonitrile (from Step B) for 3-trifluoromethyl-4-(2-(S)-methylpropyloxy) benzonifrile: iH NMR (500 Mhz) δ 1.0 (t, J= 7.3, 3H), 1.32 (d, J= 5.9, 3H), 1.68 (m, IH), 1.79 (m, IH), 4.45 (m, IH), 7.65 (d, J= 8.3, 2H).
CARBOXYLIC ACID 18 4-(2-(S -Butoxy)benzoic acid Step A: Methyl 4-(2-(S)-butoxy)benzoate
The title compound was prepared using procedure analogous to that described in CARBOXYLIC ACID 16, Step A substituting methyl 4- hydroxybenzoate for 3-fluoro-4-hydroxybenzaldehyde.
Step B: 4-(2-(S)-Butoxy)benzoic acid
A solution of 1.0 g (4.8 mmol) of methyl 4-(2-(S)-butoxy)benzoate in 15 mL of MeOH was treated with 1 mL of 5.0 N NaOH at rt for 1 h. The solution was concentrated, acidified with 6 mL of 2 N HCl , extracted with EtOAc, dried and concentrated to afford 800 mg (86%) of the title compound.
CARBOXYLIC ACID 19
4-(2-(S -Butoxy-2-fluoro-benzoic acid Step A: 4-(2-(S)-Butoxy-2-fluoro-benzonitrile
The title compound was prepared using a procedure analogous to that described in CARBOXYLIC ACID 16, Step A substituting 2-fluoro-4-hydroxy- benzonitrile for 3-fluoro-4-hydroxybenzaldehyde.
Step B: 4-(2-(S)-Butoxy-2-fluoro-benzoic acid
A mixture of 770 mg (4.0 mmol) of 4-(2-(S)-butoxy-2-fruoro- benzonitrile (from Step A) 20 mL of EtOH and 8 mL of 5 N NaOH (8 ml) was stirred at 80°C for 20 hours. The solution was concentrated, acidified with 2 N HCl, extracted with EtOAc, dried and concentrated to yield 0.57 g of the title compound: IH NMR (500 Mhz) δ 7.99 (t, J= 8.8 , IH), 6.75 (dd, J= 2.0, 6.9 , IH), 6.66 (dd, J= 2.1, 11.0 , IH), 4.38-4.44 (m, 2H), 1.75-1.85 (m, IH), 1.65-1.75 (m, IH), 1.37 (d, J= 6.0 , 3H), 1.02 (t, J= 7.4 , 3H).
CARBOXYLIC ACID 20 3,5-Difluoro-4-(2.2.2-trifluoroethoxy benzoic acid Step A: 5-Bromo-l,3-difluoro-2-(2,2,2-trifluoroethoxy)benzene
A mixture of 1.25 g (6 mmol) of 4-bromo-2,6-difluorophenol and 3.93 g (12 mmol) of cesium carbonate in 10 mL of acetonifrile was treated with 1.4 g (6 mmol) of 2,2,2-trifluoroethyltrifluoromethanesulfonate and stirred at rt for 2 h. The reaction mixture was diluted with EtOAc and washed with 2 N HCl. The organic layer was dried and concentrated. Silica gel chromatography using 9:1 hexanes/EtOAc as the eluent afforded 230 mg of the title compound: iH NMR (500 Mhz) δ 7.16 (d, J= 7.3 , 2H), 4.41-4.50 (m, 2H).
Step B: 3,5-Difluoro-4-(2,2,2-trifluoroethoxy)benzonitrile
A mixture of 230 mg (1.8 mmol) of 5-bromo-l,3-difluoro-2-(2,2,2- trifluoroethoxy)benzene (from Step A), 63 mg (1.1 mmol) of zinc cyanide, 41 mg (0.09 mmol) of tris(dibenzylideneacetone)dipalladium(0) and 60 mg (0.21 mmol) of l,r-bis(diphenylphosino)feπocene in 1.5 mL DMF and and 15uL water was heated at 95 °C for 2 h. The reaction mixture was cooled and concentrated. Silica gel chromatography using 9:1 hexanes/EtOAc as the eluant afforded 50 mg of the title compound.
Step C: 3,5-Difluoro-4-(2,2,2-trifluoroethoxy)benzoic acid
The title compound was prepared using a procedure analogous to that described in CARBOXYLIC ACID 7, Step B substituting 3,5-difluoro-4-(2,2,2- trifluoroethoxy) benzonifrile for 3-trifluoromethyl-4-(2-(S)-methylpropyloxy) benzonifrile: lH NMR (500 Mhz) δ 7.71 (d, J= 8.1 , 2H), 4.58-4.64 (m, 2H). J
CARBOXYLIC ACID 21 5-(2-Methyl-l-oxopropyl pyridine-2-carboxylic acid Step A: (+/-)-5-(2-Methyl-l-hydroxypropyl)-2-bromopyridine
A solution of 1.00 g (4.4 mmol) of 2,5-dibromopyridine in 10 mL of THF at 0 °C was treated with 2.5 mL of 2 M isopropylmagnesium chloride solution in THF and the resulting mixture was stined cold for 1 h. The mixture was treated with 0.46 mL (5.1 mmol) of isobutyraldehyde, warmed to rt and stined for 16 h. The mixture was partitioned between 50 mL of EtOAc and 50 mL of water and the layers were separated. The organic layer was washed with 25 mL of sat'd NaCl, dried and concentrated. Silica gel chromatography using 3:1 v/v hexanes/EtOAc as the eluant gave 290 mg of the title compound: H NMR (500 MHz , CDCI3) δ 8.29 (d, J= 2.3, ,
IH), 7.55 (dd, J= 2.3, 8.0 , IH), 7.47 (d, J= 8.3 , IH), 4.45 (d, J= 6.7 , 1H), 1.94 (m, 1H), 0.97 (d, J= 6.6 , 3H), 0.85 (d, J= 6.9 , 3H).
Step B: 5-(2-Methyl-l-oxopropyl)-2-bromopyridine
A mixture of 290 mg (1.25 mmol) of 5-(2-methyl-l-hydroxypropyl)-2- bromopyridine (from Step A) and 220 mg (1.9 mmol) of N-methylmorpholine-N- oxide in 5 mL of CH2CI2 was treated with 20 mg of tetrapropylarnmonium perruthenate. The mixture was stined at rt for 3 h. Silica gel chromatography of the reaction mixture using 10: 1 v/v hexanes/EtOAc as the eluant and afforded 230 mg of the title compound: lH NMR (500 MHz , CDCI3) δ 8.29 (d, J= 2.5, , IH), 8.07 (dd,
J= 2.6, 8.3 , IH), 7.61 (d, J= 8.5 , IH), 3.45 (m, IH), 1.23 (d, J= 6.8 , 6H).
Step C: 5-(2-Methyl-l-oxopropyl)pyridine-2-carbonitrile A solution of 300 mg (1.3 mmol) of 5-(2 -methyl- l-oxopropyl)-2- bromopyridine (from Step B), zinc cyanide (0.093 g, 0.789 mmol), tris(dibenzylideneacetone)-dipalladium(0) (24 mg, 0.026 mmol) and 1,1- bis(diphenylphosphino)-feπocene (33 mg, 0.059 mmol) in 2 mL of DMF and 0.03 mL of water was heated at 80 °C for 2.5 h. The reaction was cooled, loaded onto silica gel and eluted with 5:1 v/v hexanes/EtOAc to give 224 mg of the product: Η NMR (500 MHz , CDCI3) δ 9.21 (d, J= 1.8, , IH), 8.34 (dd, J= 2.3, 8.0 , IH), 7.83 (d, J= 8.0
, IH), 3.50 (m, IH), 1.25 (d, J= 6.8 , 6H). Step D: 5-(2-Methyl-l-oxopropyl)pyridine-2-carboxylic acid
A solution of 125 mg (0.7 mmol) of 5-(2-methyl-l- oxopropyl)pyridine-2-carbonitrile (from Step C) and 0.7 mL of 5.0 N NaOH in 2.5 mL of EtOH was stined at 75 °C for 1 h. The reaction was cooled, diluted with 50 mL of EtOAc, washed with 20 mL of 2 N HCl, 25 mL of sat' d NaCl, dried and concentrated to give 108 mg of the title compound.
CARBOXYLIC ACID 22 5-( 1 , 1 -Difluoro-2-methylpropyl pyridine-2-carboxylic acid The title compound was prepared from 5-(2 -methyl- 1 - oxopropyl)pyridine-2-carbonitrile (from CARBOXYLIC ACID 21, Step C) using procedures analogous to those described in CARBOXYLIC ACID 4, Steps B and C: Η NMR (500 MHz , CDCI3) δ 8.71 (s, IH), 8.30 (d, J= 8.0 , IH), 8.01 (dd, J=
2.1, 8.3 , IH), 2.37 (m, IH), 1.04 (d, J= 6.9 , 6H); ESI-MS 216.7 (M+H).
CARBOXYLIC ACID 23
(S)-4-(3,3-Difluorocyclopentyl benzoic acid Step A: (S)-3-(4-Bromophenyl)cyclopentanone
To a mixture of 7.2 g (35.8 mmol) of 4-bromophenylboronic acid, 186 mg (0.72 mmol) of acetylacetonatobis(ethylene)rhodium (I) and 446 mg (0.71 mmol) of (S)-2,2'-bis(diphenylphosphino)-l,l'binaphthyl (BLNAP) in 60 mL of dioxane and 6 mL of H2O under nitrogen was added 1.0 mL (11.9 mmol) of 2-cyclopenten-l-one.
After refluxing for 5.5 h, the reaction was concentrated. The residue was partitioned between 300 mL of EtOAc and 300 mL of 1 N NaHCO3. After separating phases, the organic layer was washed with 300 mL of brine, dried over Na2SO4 and concentrated. The residue was purified on a 40M Biotage column using 9:1 v/v hexane/EtOAc as the eluant to afford 1.90 g of the title compound as a white solid: 1H-NMR (500 MHz) δ 1.97 (m, IH), 2.29-2.37 (m, 2H), 2.43-2.52 (m, 2H), 2.69 (m, IH), 3.40 (m, IH), 7.16 (d, J = 8.5, 2H), 7.49 (d, J = 8.5, 2H).
Step B: (S)-3-(4-Bromophenyl)-l,l-difluorocyclopentane
A mixture of 2.1 mL (11.4 mmol) of [bis(2-methoxyethyl)amino] sulfur trifluoride and 0.10 mL (0.7 mmol) of borontrifluoride etherate in 7 mL of toluene at 0 °C was allowed to stand for 1.3 h with occasional stining. A solution of 1.9 g (7.9 mmol) of (S)-3-(4-bromophenyl)cyclopentanone (from Step A) in 13 mL of toluene was added. The reaction was stined at 55 °C for 2 days. After cooling, the mixture was added to 250 mL of 2N NaOH and 250 mL of Et2θ at 0 °C. After stining for 30 min, the phases were separated. The organic layer was washed with 250 mL of 1 N NaOH and 250 mL of H2O, dried over MgSO4 and concentrated. The residue was purified on a 40M Biotage column using 49:1 v/v hexane/Et2θ as the eluant to afford 1.47 g of the title compound: iH-NMR (500 MHz) δ 1.85 (m, IH), 2.09-2.26 (m, 3H), 2.35 (m, IH), 2.56 (m, IH), 3.30 (m, IH), 7.13 (d, J = 8.3, 2H), 7.46 (d, J = 8.3, 2H).
Step C: (S)-4-(3,3-Difluorocyclopentyl) benzoic acid
A solution of 1.0 g (3.8 mmol) of (S)-3-(4-bromophenyl)-l,l- difluorocyclopentane' (from Step B) in 15 mL of THF at -78 °C was treated with 1.6 mL (4.0 mmol) of 2.5M BuLi in hexanes. After stining for 15 min, the reaction was added to a suspension of dry ice in 200 mL of Et2θ. The mixture was allowed to warm to rt. The reaction mixture was extracted with 100 mL of 1 N NaOH. After separating phases, the aqueous layer was acidified to pH 1-2 with concentrated HCl. The aqueous phase was extracted with 3 x 100 mL of CH2CI2. The combined organic phases were dried and concentrated to give 0.67 g of the title compound: lH- NMR (500 MHz, CD3OD) δ 1.87 (m, IH), 2.13-2.37 (m, 4H), 2.54 (m, IH), 3.41 (m,
IH), 7.39 (d, J = 8.2, 2H), 7.97 (d, J = 8.2, 2H).
CARBOXYLIC ACID 24 (R)-4-(3.3-Difluorocvclopentyl benzoic acid
The title compound was prepared using analogous procedures to CARBOXYLIC ACID 23, except (R)-2,2'-bis(diphenylphosphino)-l,l'binaphthyl (BLNAP) was substituted for (S)-2,2'-bis(diphenylphosphino)-l, binaphthyl (BLNAP) in Step A. PREPARATION OF EXAMPLE COMPOUNDS
EXAMPLE 1 3-(2-(N-Methylamino pyridin-3-yl)-5-(4-(2-methylpropyl')phenylVl,2.4-oxadiazole Step A: 3-(2-(Chloro)pyridin-3-yl)-5-(4-(2-methylpropyl)phenyl)-l,2,4-oxadiazole A mixture of 500 mg (2.8 mmol) of 4-(2-methylpropyl)benzoic acid, 600 mg (3.1 mmol) of l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide hydrochloride and 420 mg (3.1 mmol) of 1-hydroxybenzotriazole (0.42 g, 3.09 mmol) in 10 mL of DMF was stined at rt for 10 min. N-Hydoxyamidine 1 (620 mg, 3.6 mmol) was added and the resulting mixture was heated at 120 °C for 3 h. The reaction was cooled and concentrated. Silica gel chromatography using 3:1 v/v hexanes/EtOAc as the eluant afforded 103 mg of the title compound: Η NMR (500 MHz , CDCI3) δ 8.56 (dd, J= 2.0, 4.8 , IH), 8.38 (dd, J= 2.1, 7.6 , IH), 8.12 (d, J= 8.2
, 2 H), 7.42 (dd, J= 4.8, 7.6 , IH), 7.35 (d, J= 8.2 , 2H), 2.59 (d, J- 7.1 , 2H), 1.94 (m, IH), 0.94 (d, J= 6.7 , 6H); ESI-MS 314.1 (M+H).
Step B: 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2-methylpropyl)phenyl)-l,2,4- oxadiazole
A solution of 50 mg (0.12 mmol) of 3-(2-(chloro)pyridin-3-yl)-5-(4-(2- methylpropyl) phenyl)- 1,2,4-oxadiazole (from Step A) and 0.05 mL of diethanolamine in 0.5 mL of N-methylformamide was stined at 120 °C for 16 h. The reaction was cooled and concentrated. Chromatography on silica gel using 5:1 v/v hexanes/EtOAc as the eluant afforded 20 mg of the title compound as a white solid: lH NMR (500 MHz , CDCI3) δ 8.43 (dd, J= 2.1, 7.8 , IH), 8.33 (dd, J= 1.8, 8.3 , IH), 8.12 (d, J= 8.3 , 2 H), 7.33 (d, J= 8.2 , 2H), 7.14-7.20 (bs, IH), 6.70 (dd, J= 5.0, 7.5 , IH), 3.18 (d, J= 4.6 , 3H), 2.58 (d, J= 7.1 , 2H), 1.94 (m, IH), 0.94 (d, J= 6.6 , 6H); ESI-MS 309.1 (M+H).
EXAMPLES 2-9 The following were prepared using procedures analogous to those described in EXAMPLE 1 substituting 4-(cyclohexyl)benzoic acid for 4-(2- methylpropyl)benzoic acid and the appropriate N-HYDROXYAMIDINE for N- HYDROXYAMIDINE 1 in Step A and the appropriate amine for N- methylformamide in Step B.
Figure imgf000069_0001
Figure imgf000069_0002
Figure imgf000070_0001
EXAMPLES 10-13 The following were prepared using procedures analogous to those described in EXAMPLE 1 substituting the appropriate CARBOXYLIC ACID for 4- (2-methylρropyl) benzoic acid and N-HYDROXYAMIDINE 3 for N- HYDROXYAMIDINE 1 in Step A
Figure imgf000071_0001
Figure imgf000071_0002
Figure imgf000072_0001
EXAMPLES 14-17 The following were prepared using procedures analogous to those described in EXAMPLE 1 substituting the appropriate CARBOXYLIC ACID for 4- (2-methylpropyl) benzoic acid in Step A and the appropriate amine for N- methylformamide in Step B.
Figure imgf000072_0002
Figure imgf000073_0001
EXAMPLE 19 3 -(2-(N-Methylamino)pyridin-3 -yl -5-(4-(2.2-difluoropropyl phenvD- 1.2.4- oxadiazole A mixture of 50 mg (0.25 mmol) of 4-(2,2-difluoropropyl)benzoic acid, 50 mg (0.3 mmol) of N-HYDROXYAMIDINE 1 and 72 mg (0.37 mmol) of 1- [3-(dimethylamino) propyl]-3-ethylcarbodiimide hydrochloride and in 1 mL of 1,2- dichloroethane was stined at rt for 6 h , then at 80 °C for 16 h. The reaction was cooled and concentrated. Silica gel chromatography using 10: 1 v/v hexanes/EtOAc as the eluant afforded 19 mg of the title compound: Η NMR (500 MHz , CDCI3) δ
8.45 (d, J= 6.7 , IH), 8.34 (dd, J= 1.9, 4.8 , IH), 8.18 (d, J= 8.2 , 2 H), 7.48 (d, J= 8.3 , 2H), 6.72 (dd, J= 4.8, 7.6 , IH), 3.20-3.30 (m, 5H), 1.60 (d, J= 18.3 , 3H) ; ESI-MS 331.3 (M+H).
EXAMPLES 20-46 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl) benzoic acid and the appropriate N-HYDROXYAMIDINE for N- HYDROXYAMIDINE 1.
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
EXAMPLES 47-56 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate CARBOXYLIC ACID for 4- (2,2-difluoropropyl) benzoic acid and N-HYDROXYAMIDLNE 7 for N-
HYDROXYAMIDINE 1.
Figure imgf000079_0002
Figure imgf000079_0003
Figure imgf000080_0001
Figure imgf000081_0001
EXAMPLE 58 3-(2-(N-Methylamino)pyridin-3-yl)-5-(5-(2-methylpropyl)pyridin-2-yl - 2,4- oxadiazole Step A: 3-(2-(Benzotriazol-l-yloxy)pyridin-3-yl)-5-(5-bromopyridin-2-yl)-l,2,4- oxadiazole A solution of 300 mg (1.45 mmol) of 5-bromopyridine-2- CARBOXYLIC ACID, 310 mg (1.6 mmol) of l-[3-(dimethylamino)propyl]-3- ethylcarbodiimide hydrochloride and 220 mg (1.6 mmol) of 1-hydroxybenzotriazole (0.22 g, 1.64 mmol) in 2 mL of DMF was stined at rt for 2 h. The mixture was treated with 310 mg (1.8 mmol) of N-HYDROXYAMIDINE 1 then stined at rt for 1 h and 80 °C for 16 h. The reaction was cooled and concentrated. Silica gel chromatography using 1:1 v/v hexanes/EtOAc as the eluant gave 190 mg of the title compound: ESI-MS 437.9 (M+H).
Step B: 3-(2-(Benzotriazol-l-yloxy)ρyridin-3-yl)-5-(5-(2- methylpropyl)pyridin-2-yl)- 1 ,2,4-oxadiazole
A solution of 190 mg (0.43 mmol) of 3-(2-(benzotriazol-l- yloxy)pyridin-3-yl)-5-(5-bromopyridin-2-yl)- 1,2,4-oxadiazole (from Step A) in 1.0 mL of 0.M isobutylzinc bromide solution in THF was treated with ~2 mg of bis(tri-t- butylphosphine) palladium(O) and the resulting mixture was stined at rt for 2 h.
Silica gel chromatography using 3:2 v/v hexanes/EtOAc gave the title compound:
ESI-MS 414.1 (M+H).
Step C: 3-(2-(N-Methylamino)pyridin-3-yl)-5-(5-(2-methylpropyl)pyridin-2- yl)- 1,2,4-oxadiazole
A mixture of 100 mg (0.24 mmol) of 3-(2-(benzotriazol-l- yloxy)pyridin-3-yl)-5-(5-(2-methylpropyl)pyridin-2-yl)-l ,2,4-oxadiazole (from Step B), 100 mg of diethanolamine in 0.5 mL of N-methylformamide (0.5 ml) was stined at 130°C for 16 h. The reaction was cooled and concentrated. The title compound was after purification by HPLC B: iH NMR (500 MHz , CDCI3) δ 8.91 (dd, J= 1.3,
7.5 , IH), 8.70 (bs, IH), 8.48 (d, J= 6.2 , IH), 8.43 (bs, IH), 8.25 (d, J= 8.0 , IH), 7.77 (dd, J= 1.6, 8.1 , IH), 7.02 (t, J= 6.2 , IH), 3.44 (s, 3H), 2.65 (d, J= 7.1 , 2H), 1.97 (m, IH), 0.98 (d, J= 6.6 , 6H); ESI-MS 310.2 (M+H).
EXAMPLE 59
3-(2-(N-Methylamino pyridin-3-yl)-5-(4-bromophenyl -1.2.4-oxadiazole
A solution of 5.0 g (31.6 mmol) of N-HYDROXYAMIDINE 7 and 4.6 mL (33.1 mmol) of triethylamine in 50 mL of DMF at 0 °C was treated with 6.9 g (31.6 mmol) of 4-bromobenzoyl chloride. The reaction was stined at 0 ° for 1 hour, then heated to 120°C for 2 hours. The reaction was cooled, diluted with methanol (50 ml) and the product collected by filtration (3.1 g): Η NMR (500 MHz , CDCI3) δ 8.41 (dd, J= 1.8, 7.6 , IH), 8.34 (dd, J= 1.9, 4.8 , IH), 8.08 (d, J= 8.5 , 2H), 7.71 (d, J= 8.7 , 2H), 7.08-7.14 (bs, IH), 6.70 (dd, J= 4.8, 7.5 , IH), 3.18 (d, J= 4.8 , 3H); ESI- MS 333.1 (M+H).
EXAMPLE 60 3-(2-(N-Methylamino ρyridin-3-ylV5-(4-(2.2.2-trifluoroethoxy phenylV1.2.4- oxadiazole
Step A: 3-(2-(Chloro)pyridin-3-yl)-5-(4-hydroxyphenyl)- 1 ,2,4-oxadiazole
The title compound was prepared using a procedure analogous to that described in EXAMPLE 1, Step A substituting 4-hydroxybenzoic acid for 4-(2- methylpropyl)benzoic acid.
Step B: 3-(2-(Chloro)pyridin-3-yl)-5-(4-(2,2,2-trifluoroethoxy)phenyl)-l,2,4- oxadiazole
A mixture of 35 mg of 3-(2-(chloro)pyridin-3-yl)-5-(4- hydroxyphenyl)- 1,2,4-oxadiazole (from Step A) and 210 mg (0.38 mmol) of cesium carbonate in 0.7 mL of acetonifrile and 0.3 mL of THF was treated with 90 mg (0.38 mmol) of 2,2,2-trifluoroethoxy trifluoromethanesulfonate. The resulting mixture was stined at rt for 2 h. Silica gel chromatography using 3:1 v/v hexanes/EtOAc as the eluant afforded 15 mg the title compound: ESI-MS 356.1 (M+H).
Step C: 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2,2,2-trifluoroethoxy)phenyl)- 1,2,4-oxadiazole
A mixture of 15 mg of 3-(2-(chloro)ρyridin-3-yl)-5-(4-(2,2,2- trifluoroethoxy) phenyl)- 1,2,4-oxadiazole (from Step B), 0.11 mL of 2 M methylamine solution in THF and Methyl amine (2.0M in THF) (0.1 lmL, 0.21 mmol) and 0.037 mL (0.21 mmol) was stined at 65 °C for 48 h. The reaction was cooled and concentrated. Silica gel chromatography using 8:1 hexanes/EtOAc as the eluant afforded 6.2 mg of the title compound: Η NMR (500 MHz , CDCI3) δ 8.55 (s, IH), 8.39 (d, J= 4.1 , IH), 8.24 (d, J= 8.5 , 2H), 7.16 (d, J= 8.5 , 2H), 6.82 (s, IH), 4.46- 4.54 (m, 2H), 3.33 (s, 3H); ESI-MS 351.1 (M+H)
EXAMPLE 60a 3-(2-(N-Methylamino pyridin-3-yl -5-(4-(2-fluoro-l-fluoromethyl ethoxy-3- trifluoromethylphenyl - 1 ,2,4-oxadiazole Step A: 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-fluoro-3-trifluoromethylphenyl)- 1,2,4-oxadiazole
A mixture of 200 mg of N-HYDROXYAMIDINE 7 and 0.44 mL (3.1 mmol) of TEA in 0.4 mL of toluene and 1.4 mL of DMF at 0 °C was treated with 360 mg (1.6 mmol) of 3-trifluoromethyl-4-fluoro benzoyl chloride. The resulting mixture was stined at 120 °C for 2.5 h. The mixture was cooled, then partitioned between CH2CI2 and water. The organic layer was separated, dried and concentrated. Silica gel chromatography using 4:1 v/v hexanes/EtOAc afforded 150 mg of the title compound: ESI-MS 340.2 (M+H).
Step B: 3-(2-(N-Methylamino)pyridin-3-yl)-5-(4-(2-fluoro-l-fluoromethyl)ethoxy-3- trifluoromethylphenyl) - 1 ,2,4-oxadiazole
The title compound was prepared from 3-(2-(N-methylamino)pyridin- 3-yl)-5-(4-fluoro-3-trifluoromethylphenyl)-l,2,4-oxadiazole (from Step A) using a procedure analogous to that described in CARBOXYLIC ACID 7, Step A, substituting l,3-difluoro-2-propanol for 2-(S)-butanol: Η NMR (500 MHz , CDCI3) δ 8.51 (s, IH), 8.41 (d, J= 7.1 , 3H), 7.37 (d, J= 8.7 , 2H), 6.80 (s, IH), 4.95-5.09 (m, IH), 4.84 (s, 2H), 4.75 (s, 2H), 3.21-3.30 (m, 3H); ESI-MS 415.2 (M+H).
EXAMPLE 61
3-(2-(N-methylamino pyridin-3-yl -5-(4-(3-thienyl phenyl -l,2,4-oxadiazole A solution of 50 mg (0.15 mmol) of 3-(2-(N-methylamino)pyridin-3- yl)-5-(4-bromophenyl)- 1,2,4-oxadiazole (from EXAMPLE 61), 29 mg (0.23 mmol) of 3-thiopheneboronic acid and 26 mg (0.45 mmol) potassium fluoride in 1 mL of THF was treated with 7 mg (0.003 mmol) of palladium(II) acetate and 2.1 mg (0.006 mmol) of 2-(dicyclohexylphosphino)biphenyl. The resulting mixture was stined at 50 °C for 2 h, then cooled and concentrated. Silica gel chromatography using 7: 1 v/v hexanes/EtOAc as the eluant afforded 30 mg of the title compound: Η NMR (500 MHz , CDCI3) δ 8.44 (dd, J= 1.9, 7.8 , IH), 8.34 (dd, J= 1.8, 4.8 , IH), 8.23 (d, J= 8.5
, 2H), 7.78 (d, J= 8.5 , 2H), 7.63 (dd, J= 1.4, 2.7 , IH), 7.45-7.48 (m, 2H), 7.16-7.20 (bs, IH), 6.71 (dd, J= 5.0, 7.8 , IH), 3.20 (d, J= 4.8 , 3H); ESI-MS 335.2 (M+H).
EXAMPLES 62-71 The following were prepared using procedures analogous to those described in EXAMPLE 61 substituting the appropriate aryl boronic acid for thiophene-3-boronic acid.
Figure imgf000085_0001
Figure imgf000085_0002
Figure imgf000086_0001
EXAMPLE 72
3-(2-(N-Methylamino pyridin-3-yl -5-(4-(2-methylpropyπ-3-
(frifluoromethyl)phenvT)- 1 ,2,4-oxadiazole
A mixture of 30 mg (0.085 mmol) of 3-(2-(N-methylamino)pyridin-3- yl)-5-(4-chloro-3-(trifluoromethyl)phenyl)- 1,2,4-oxadiazole (from EXAMPLE 24) in 1.0 mL of 0.5 M isobutylzinc bromide solution in THF) was treated with 2 mg of bis(tri-t-butylphosphine)palladium(0) (2 crystals). The resulting mixture was stined at rt for 20 h, then concentrated. Silica gel chromatography using 9:1 hexanes/EtOAc afforded 2.5 mg of title compound: Η NMR (500 MHz , CDCI3) δ 8.51 (s, IH), 8.47
(d, J= 6.4 , IH), 8.38 (s, IH), 8.31 (d, J= 7.3 , IH), 7.57 (d, J= 7.8 , IH), 7.14 (s, IH), 3.23 (s, 3H), 2.80 (d, J= 6.6 , 2H), 2.00-2.10 (m, IH), 1.01 (d, J= 6.2 , 6H); ESI-MS 377.3 (M+H).
EXAMPLES 73-80 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
Figure imgf000087_0001
Figure imgf000087_0002
Figure imgf000088_0001
EXAMPLES 81-87 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
Figure imgf000089_0001
Figure imgf000090_0001
EXAMPLES 88-90 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE l and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
Figure imgf000091_0001
Figure imgf000091_0002
EXAMPLE 91 3-(4-Aminopyrimidin-5-yl -5-(4-cvclohexylphenyl -l,2,4-oxadiazole
The title compound was prepared using procedures analogous to those described in EXAMPLE 19 substituting N-hydroxy (4-aminopyrimidin-5-yl)amidine for N-HYDROXYAMIDINE 1 and the appropriate 4-cyclohexylbenzoic acid for 4- (2,2-difluoropropyl)benzoic acid: lH NMR (CD3OD) δ 1.31-1.56, (m, 5 H), 1.77-
1.90 (m, 5 H), 2.66 (t, 1 H, J= 5.8 Hz), 7.49 (d, 2 H, J= 8.2 Hz), 8.16, (d, 2H, J= 8.2 Hz), 8.69, (s, 1 H), 9.10 (s, 1 H); ESI-MS 322 (M+H).
EXAMPLES 92-102 The following were prepared using procedures analogous to those described in
EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-
HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000093_0001
Figure imgf000094_0002
EXAMPLES 103-106 The following were prepared using procedures analogous to those described in EXAMPLE 19 substituting the appropriate N-HYDROXYAMIDINE for N-HYDROXYAMIDINE 1 and the appropriate CARBOXYLIC ACID for 4-(2,2- difluoropropyl)benzoic acid.
Figure imgf000094_0001
Figure imgf000094_0003
Figure imgf000095_0001
EXAMPLE 107 3-(2-Amino-5-fluoropyridin-3-yl)-5-(3-trifluoromethyl-4-(l,l.l-trifluoro-2-(S)- propyloxy))phenyl)- 1 ,2,4-oxadiazole To a mixture of CARBOXYLIC ACID 15 (53 mg, 0.176 mmol) in acetonifrile (1.0 mL), EDC-HC1 (34 mg, 0.176 mmol) was added. After 30 min, the resultant solution was added to a mixture of N-HYDROXYAMIDINE 11 and acetonifrile (1.0 mL) in a sealed tube and heated to 40 °C. After 4 hr, the reaction mixture was heated to 120 °C for 20 hr. The reaction mixture was cooled to ambient temperature, concentrated in vacuo and purified by flash chromatography (10, 15% EtOAc/hexanes) on SiO2 to afford 40 mg of the title compound as a white film. This material was further purified by HPLC. Conditions: Chiralcel OD 4.6 x 250 mm column, 60:40 v/v heptane/tPrOH, 1.0 mL/min, λ = 210 nM. (R)-enantiomer = 12.6 min, (S)-enantiomer = 13.7 min: 1H NMR (500 MHz , CDC13) δ 1.61 (d, 3 H, J= 6.4 Hz), 4.91 (septet, 1 H, J= 6.1 Hz), 6.06 (br, 2 H), 7.22 (d, 1 H, J= 8.9 Hz), 8.13, (d, 1 H, J= 3.0 Hz), 8.20 (dd, 1 H, J= 3.0, 8.7 Hz), 8.37 (d, 1 H, J= 2.0, 8.7 Hz), 8.48 (d, 1 H, J= 2.0 Hz); HPLC/MS (HPLC A): 437 (M+H)+, 3.89 min.
EXAMPLE 108 3-(2-(TS-Methylamino)-5-fluoropyridin-3-yl)-5-(3-trifluoromethyl-4-(l,l,l-trifluoro- 2-(S)-propyloxy) phenyl)- 1 ,2,4-oxadiazole The title compound was prepared using a procedure analogous to that described for EXAMPLE 107 substituting N-HYDROXYAMIDINE 10 for N-
HYDROXYAMIDINE 11 : 1H NMR (500 MHz , CDC13) δ 1.62 (d, 3 H, J= 6.6 Hz), 3.15 (d, 3 H, J= 4.8 Hz), 4.91 (septet, 1 H, J= 6.0 Hz), 6.95 (d, 1 H, J= 4.1 Hz), 7.21, (d, 1 H, J= 8.9 Hz), 8.19 (dd, J= 3.0, 8.7 Hz), 8.22 (d, 1 H, J= 2.9 Hz), 8.36 (dd, 1 H, J= 2.1, 8.7 Hz), 8.47 (d, 1 H, J= 2.1 Hz); HPLC/MS (HPLC A): 451 (M+H)+, 4.18 min.
BIOLOGICAL ACTIVITY
The SlPi/Edgl, SlP3,/Edg3, SlP2/Edg5, SlP4/Edg6 or SIP5 /Edg8 activity of the compounds of the present invention can be evaluated using the following assays:
Ligand Binding to Edg/SIP Receptors Assay 33p-sphingosine- 1 -phosphate was synthesized enzymatically from γ33p_ATP and sphingosine using a crude yeast extract with sphingosine kinase activity in a reaction mix containing 50 mM KH2PO4, 1 mM mercaptoethanol, 1 mM Na3VO4, 25 mM KF, 2 mM semicarbazide, 1 mM Na2EDTA, 5 mM MgCl2, 50 mM sphingosine, 0.1% TritonX-114, and 1 mCi γ33p_ATP (NEN; specific activity 3000 Ci/mmol). Reaction products were extracted with butanol and 33p_Sphingosine-l- phosphate was purified by HPLC. Cells expressing EDG/S1P receptors were harvested with enzyme-free dissociation solution (Specialty Media, Lavallette, NJ). They were washed once in cold PBS and suspended in binding assay buffer consisting of 50 mM HEPES-Na, pH 7.5, 5mM MgCl2, lmM CaCl2, and 0.5% fatty acid-free BSA. 33p-sphfngosine-l- phosphate was sonicated with 0.1 nM sphingosine- 1-phosphate in binding assay buffer; 100 μl of the ligand mixture was added to 100 μl cells (1 x 106 cells/ml) in a 96 well microtiter dish. Binding was performed for 60 min at room temperature with gentle mixing. Cells were then collected onto GF/B filter plates with a Packard Filtermate Universal Harvester. After drying the filter plates for 30 min, 40 μl of Microscint 20 was added to each well and binding was measured on a Wallac
Microbeta Scintillation Counter. Non-specific binding was defined as the amount of radioactivity remaining in the presence of 0.5 μM cold sphingosine- 1-phosphate.
Alternatively, ligand binding assays were performed on membranes prepared from cells expressing Edg/SIP receptors. Cells were harvested with enzyme-free dissociation solution and washed once in cold PBS. Cells were disrupted by homogenization in ice cold 20 mM HEPES pH 7.4, 10 mM EDTA using a Kinematica polytron (setting 5, for 10 seconds). Homogenates were centrifuged at 48,000 x g for 15 min at 40C and the pellet was suspended in 20 mM HEPES pH 7.4, 0.1 mM EDTA. Following a second centrifugation, the final pellet was suspended in 20 mM HEPES pH 7.4, 100 mM NaCl, 10 mM MgCl2- Ligand binding assays were performed as described above, using 0.5 to 2 μg of membrane protein.
Agonists and antagonists of Edg/SIP receptors can be identified in the 33p-sphingosine- 1-phosphate binding assay. Compounds diluted in DMSO, methanol, or other solvent, were mixed with probe containing 33p_sphingosine-l- phosphate and binding assay buffer in microtiter dishes. Membranes prepared from cells expressing Edg/SIP receptors were added, and binding to 3p_sphingosine-l- phosphate was performed as described. Determination of the amount of binding in the presence of varying concentrations of compound and analysis of the data by nonlinear regression software such as MRLCalc (Merck Research Laboratories) or PRISM (GraphPad Software) was used to measure the affinity of compounds for the receptor. Selectivity of compounds for Edg/SIP receptors was determined by measuring the level of 33p_sphingosine- 1-phosphate binding in the presence of the compound using membranes prepared from cells transfected with each respective receptor (SlPi/Edgl, SlP3/Edg3, SlP2/Edg5, SlP4/Edg6, SlPs/Edg8).
35s-GTPγS Binding Assay
Functional coupling of SlP/Edg receptors to G proteins was measured in a 5s-GTPγS binding assay. Membranes prepared as described in the Ligand Binding to Edg/SIP Receptors Assay (1-10 μg of membrane protein) were incubated in a 200 μl volume containing 20 mM HEPES pH 7.4, 100 mM NaCl, 10 mM MgC-2,
5 μM GDP, 0.1% fatty acid-free BSA (Sigma, catalog A8806), various concentrations of sphingosine- 1-phosphate, and 125 pM 35S-GTPγS (NEN; specific activity 1250 Ci/mmol) in 96 well microtiter dishes. Binding was performed for 1 hour at room temperature with gentle mixing, and terminated by harvesting the membranes onto GF/B filter plates with a Packard Filtermate Universal Harvester. After drying the filter plates for 30 min, 40 μl of Microscint 20 was added to each well and binding was measured on a Wallac Microbeta Scintillation Counter.
Agonists and antagonists of SlP/Edg receptors can be discriminated in the 35s-GTPγS binding assay. Compounds diluted in DMSO, methanol, or other solvent, were added to microtiter dishes to provide final assay concentrations of 0.01 nM to 10 μM. Membranes prepared from cells expressing SlP/Edg receptors were added, and binding to 5s-GTPγS was performed as described. When assayed in the absence of the natural ligand or other known agonist, compounds that stimulate 35 s- GTPγS binding above the endogenous level were considered agonists, while compounds that inhibit the endogenous level of 35s-GTPγS binding were considered inverse agonists. Antagonists were detected in a 35s-GTPγS binding assay in the presence of a sub-maximal level of natural ligand or known SlP/Edg receptor agonist, where the compounds reduced the level of 35s-GTPγS binding. Determination of the amount of binding in the presence of varying concentrations of compound was used to measure the potency of compounds as agonists, inverse agonists, or antagonists of SlP/Edg receptors. To evaluate agonists, percent stimulation over basal was calculated as binding in the presence of compound divided by binding in the absence of ligand, multiplied by 100. Dose response curves were plotted using a non-linear regression curve fitting program MRLCalc (Merck Research Laboratories), and EC50 values were defined to be the concentration of agonist required to give 50% of its own maximal stimulation. Selectivity of compounds for SlP/Edg receptors was determined by measuring the level of 35S-GTPγS binding in the presence of compound using membranes prepared from cells transfected with each respective receptor.
Intracellular Calcium Flux Assay
Functional coupling of SlP/Edg receptors to G protein associated intracellular calcium mobilization was measured using FLIPR (Fluorescence Imaging Plate Reader, Molecular Devices). Cells expressing SlP/Edg receptors were harvested and washed once with assay buffer (Hanks Buffered Saline Solution (BRL) containing 20mM HEPES, 0.1% BSA and 710 μg/ml probenicid (Sigma)). Cells were labeled in the same buffer containing 500 nM of the calcium sensitive dye Fluo- 4 (Molecular Probes) for 1 hour at 37°C and 5% CO2. The cells were washed twice with buffer before plating 1.5x105 per well (90μl) in 96 well polylysine coated black microtiter dishes. A 96-well ligand plate was prepared by diluting sphingosine- 1- phosphate or other agonists into 200 μl of assay buffer to give a concentration that was 2-fold the final test concentration. The ligand plate and the cell plate were loaded into the FLIPR instrument for analysis. Plates were equilibrated to 37°C. The assay was initiated by transfening an equal volume of ligand to the cell plate and the calcium flux was recorded over a 3 min interval. Cellular response was quantitated as area (sum) or maximal peak height (max). Agonists were evaluated in the absence of natural ligand by dilution of compounds into the appropriate solvent and transfer to the Fluo-4 labeled cells. Antagonists were evaluated by pretreating Fluo-4 labeled cells with varying concentrations of compounds for 15 min prior to the initiation of calcium flux by addition of the natural ligand or other SlP/Edg receptor agonist.
Preparation of Cells Expressing SlP/Edg Receptors
Any of a variety of procedures may be used to clone SlPi/Edgl, SlP3/Edg3, SlP2/Edg5, SlP4/Edg6 or SlP5/Edg8. These methods include, but are not limited to, (1) a RACE PCR cloning technique (Frohman, et al., 1988, Proc. Natl. Acad. Sci. USA 85: 8998-9002). 5' and/or 3' RACE may be performed to generate a full-length cDNA sequence; (2) direct functional expression of the Edg/SIP cDNA following the construction of an SlP/Edg-containing cDNA library in an appropriate expression vector system; (3) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a labeled degenerate oligonucleotide probe designed from the amino acid sequence of the SlP/Edg protein; (4) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA encoding the SlP/Edg protein. This partial cDNA is obtained by the specific PCR amplification of SlP/Edg DNA fragments through the design of degenerate oligonucleotide primers from the amino acid sequence known for other proteins which are related to the SlP/Edg protein; (5) screening an SlP/Edg-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA or oligonucleotide with homology to a mammalian SlP/Edg protein. This strategy may also involve using gene-specific oligonucleotide primers for PCR amplification of SlP/Edg cDNA; or (6) designing 5' and 3' gene specific oligonucleotides using the SlP/Edg nucleotide sequence as a template so that either the full-length cDNA may be generated by known RACE techniques, or a portion of the coding region may be generated by these same known RACE techniques to generate and isolate a portion of the coding region to use as a probe to screen one of numerous types of cDNA and/or genomic libraries in order to isolate a full-length version of the nucleotide sequence encoding SlP/Edg.
It is readily apparent to those skilled in the art that other types of libraries, as well as libraries constructed from other cell types-or species types, may be useful for isolating an SlP/Edg-encoding DNA or an SlP/Edg homologue. Other types of libraries include, but are not limited to, cDNA libraries derived from other cells.
It is readily apparent to those skilled in the art that suitable cDNA libraries may be prepared from cells or cell lines which have SlP/Edg activity. The selection of cells or cell lines for use in preparing a cDNA library to isolate a cDNA encoding SlP/Edg may be done by first measuring cell-associated SlP/Edg activity using any known assay available for such a purpose.
Preparation of cDNA libraries can be performed by standard techniques well known in the art. Well known cDNA library construction techniques can be found for example, in Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Complementary DNA libraries may also be obtained from numerous commercial sources, including but not limited to Clontech Laboratories, Inc. and Stratagene.
An expression vector containing DNA encoding an SlP/Edg-like protein may be used for expression of S lP/Edg in a recombinant host cell. Such recombinant host cells can be cultured under suitable conditions to produce SlP/Edg or a biologically equivalent form. Expression vectors may include, but are not limited to, cloning vectors, modified cloning vectors, specifically designed plasmids or viruses. Commercially available mammalian expression vectors may be suitable for recombinant S 1 P/Edg expression.
Recombinant host cells may be prokaryotic or eukaryotic, including but not limited to, bacteria such as E. coli, fungal cells such as yeast, mammalian cells including, but not limited to, cell lines of bovine, porcine, monkey and rodent origin; and insect cells including but not limited to Drosophila and silkworm derived cell lines.
The nucleotide sequences for the various SlP/Edg receptors are known in the art. See, for example, the following: SlPj/Edgl Human
Hla, T. and T. Maciag 1990 An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to G-protein coupled receptors. J. Biol Chem. 265:9308-9313, hereby incorporated by reference in its entirety.
WO91/15583, published on October 17, 1991, hereby incorporated by reference in its entirety. WO99/46277, published on September 16, 1999, hereby incorporated by reference in its entirety. SlPj/Edgl Mouse
WO0059529, published October 12, 2000, hereby incorporated by reference in its entirety. U.S. No. 6,323,333, granted November 27, 2001, hereby incorporated by reference in its entirety. SlPj/Edgl Rat
Lado, D.C., C. S. Browe, A.A. Gaskin, J. M. Borden, and A. J. MacLennan. 1994 Cloning of the rat edg-1 immediate-early gene: expression pattern suggests diverse functions. Gene 149: 331-336, hereby incorporated by reference in its entirety.
U.S. No. 5,585,476, granted December 17, 1996, hereby incorporated by reference in its entirety.
U.S. No. 5856,443, granted January 5, 1999, hereby incorporated by reference in its entirety.
SlP3_/Edg3 Human
An, S., T. Bleu, W. Huang, O.G. Hallmark, S. R. Coughlin, E.J. Goetzl 1997 Identification of cDNAs encoding two G protein-coupled receptors for lysosphingolipids FEBS Lett. 417:279-282, hereby incorporated by reference in its entirety.
WO 99/60019, published November 25, 1999, hereby incorporated by reference in its entirety.
U.S. No. 6,130,067, granted October 10, 2000, hereby incorporated by reference in its entirety.
SlP3/Edg3 Mouse
WO 01/11022, published February 15, 2001, hereby incorporated by reference in its entirety.
SlPVEdg3 Rat
WO 01/27137, published April 19, 2001, hereby incorporated by reference in its entirety.
SlP?/Edg5 Human An, S., Y. Zheng, T. Bleu 2000 Sphingosine 1 -Phosphate-induced cell proliferation, survival, and related signaling events mediated by G Protein-coupled receptors Edg3 and Edg5. J. Biol. Chem 275: 288-296, hereby incorporated by reference in its entirety.
WO 99/35259, published July 15, 1999, hereby incorporated by reference in its entirety. WO99/54351 , published October 28, 1999, hereby incorporated by reference in its entirety.
WO 00/56135, published September 28, 2000, hereby incorporated by reference in its entirety.
SlP?/Edg5 Mouse
WO 00/60056, published October 12, 2000, hereby incorporated by reference in its entirety.
SlP?/Edg5 Rat Okazaki, H., N. Ishizaka, T. Sakurai, K. Kurokawa, K. Goto, M.
Kumada, Y. Takuwa 1993 Molecular cloning of a novel putative G protein-coupled receptor expressed in the cardiovascular system. Biochem. Biophys. Res. Comm.
190:1104-1109, hereby incorporated by reference in its entirety.
MacLennan, A.J., C. S. Browe, A.A. Gaskin, D.C. Lado, G. Shaw 1994 Cloning and characterization of a putative G-protein coupled receptor potentially involved in development. Mol. Cell. Neurosci. 5: 201-209, hereby incorporated by reference in its entirety.
U.S. No. 5,585,476, granted December 17, 1996, hereby incorporated by reference in its entirety. U.S. No. 5856,443, granted January 5, 1999, hereby incorporated by reference in its entirety.
SlP4/Edg6 Human
Graler, M.H., G. Bernhardt, M. Lipp 1998 EDG6, a novel G-protein- coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue. Genomics 53: 164-169, hereby incorporated by reference in its entirety. WO 98/48016, published October 29, 1998, hereby incorporated by reference in its entirety.
U.S. No. 5,912,144, granted June 15, 1999, hereby incorporated by reference in its entirety. WO 98/50549, published November 12, 1998, hereby incorporated by reference in its entirety.
U.S. No. 6,060,272, granted May 9, 2000, hereby incorporated by reference in its entirety.
WO 99/35106, published July 15, 1999, hereby incorporated by reference in its entirety.
WO 00/15784, published March 23, 2000, hereby incorporated by reference in its entirety.
WO 00/14233, published March 16, 2000, hereby incorporated by reference in its entirety.
SlP4/Edg6 Mouse
WO 00/15784, published March 23, 2000, hereby incorporated by reference in its entirety.
SIP Edgδ Human
Im, D.-S., J. Clemens, T.L. Macdonald, K.R. Lynch 2001 Characterization of the human and mouse sphingosine 1-phosphate receptor, SIP5
(Edg-8): Structure- Activity relationship of sphingosine 1-phosphate receptors. Biochemistry 40:14053-14060, hereby incorporated by reference in its entirety. WO 00/11166, published March 2, 2000, hereby incorporated by reference in its entirety.
WO 00/31258, published June 2, 2000, hereby incorporated by reference in its entirety.
WO 01/04139, published January 18, 2001, hereby incorporated by reference in its entirety.
EP 1 090 925, published April 11, 2001, hereby incorporated by reference in its entirety. SlP5/Edg8 Rat
Im, D.-S., C.E. Heise, N. Ancellin, B. F. O'Dowd, G.-J. Shei, R. P. Heavens, M. R. Rigby, T. Hla, S. Mandala, G. McAllister, S.R. George, K.R. Lynch 2000 Characterization of a novel sphingosine 1-phosphate receptor, Edg-8. J. Biol. Chem. 275: 14281-14286, hereby incorporated by reference in its entirety.
WO 01/05829, published January 25, 2001, hereby incorporated by reference in its entirety.
Measurement of cardiovascular effects The effects of compounds of the present invention on cardiovascular parameters can be evaluated by the following procedure:
Adult male rats (approx. 350 g body weight) were instrumented with femoral arterial and venous catheters for measurement of arterial pressure and intravenous compound administration, respectively. Animals were anesthetized with Nembutal (55 mg/kg, ip). Blood pressure and heart rate were recorded on the Gould Po-Ne-Mah data acquisition system. Heart rate was derived from the arterial pulse wave. Following an acclimation period, a baseline reading was taken (approximately 20 minutes) and the data averaged. Compound was administered intravenously (either bolus injection of approximately 5 seconds or infusion of 15 minutes duration), and data were recorded every 1 minute for 60 minutes post compound administration.
Data are calculated as either the peak change in heart rate or mean arterial pressure or are calculated as the area under the curve for changes in heart rate or blood pressure versus time. Data are expressed as mean + SEM. A one-tailed Student's paired t-test is used for statistical comparison to baseline values and considered significant at p<0.05.
The SIP effects on the rat cardiovascular system are described in Sugiyama, A., N.N. Aye, Y. Yatomi, Y. Ozaki, K. Hashimoto 2000 Effects of Sphingosine- 1 -Phosphate, a naturally occuning biologically active lysophospholipid, on the rat cardiovascular system. Jpn. J. Pharmacol. 82: 338-342, hereby incorporated by reference in its entirety. Measurement of Mouse Acute Toxicity
A single mouse is dosed intravenously (tail vein) with 0.1 ml of test compound dissolved in a non-toxic vehicle and is observed for signs of toxicity. Severe signs may include death, seizure, paralysis or unconciousness. Milder signs are also noted and may include ataxia, labored breathing, ruffling or reduced activity relative to normal. Upon noting signs, the dosing solution is diluted in the same vehicle. The diluted dose is administered in the same fashion to a second mouse and is likewise observed for signs. The process is repeated until a dose is reached that produces no signs. This is considered the estimated no-effect level. An additional mouse is dosed at this level to confirm the absence of signs.
Assessment of Lymphopenia
Compounds are administered as described in Measurement of Mouse Acute Toxicity and lymphopenia is assessed in mice at three hours post dose as follows. After rendering a mouse unconscious by CO2 to effect, the chest is opened,
0.5 ml of blood is withdrawn via direct cardiac puncture, blood is immediately stabilized with EDTA and hematology is evaluated using a clinical hematology autoanalyzer calibrated for performing murine differential counts (H2000, CARESIDE, Culver City CA). Reduction in lymphocytes by test treatment is established by comparison of hematological parameters of three mice versus three vehicle treated mice. The dose used for this evaluation is determined by tolerability using a modification of the dilution method above. For this purpose, no-effect is desirable, mild effects are acceptable and severely toxic doses are serially diluted to levels that produce only mild effects.
In Vitro Activity of Examples
The examples disclosed herein have utility as immunoregulatory agents as demonstrated by their activity as potent and selective agonists of the SlPi/Edgl receptor over the S1PR3/Edg3 receptor as measured in the assays described above. In particular, the examples disclosed herein possess a selectivity for the SlPi/Edgl receptor over the S1PR3/Edg3 receptor of more than 100 fold as measured by the ratio of EC50 for the SlPi/Edgl receptor to the EC50 for the SlP3/Edg3 receptor as evaluated in the 35s-GTPγS binding assay described above and possess an EC50 for binding to the SlPi/Edgl receptor of less than 50 nM as evaluated by the 35s-GTPγS binding assay described above.

Claims

WHAT IS CLAIMED IS:
1. A compound represented by Formula I
Figure imgf000108_0001
I or a phannaceutically acceptable salt thereof, wherein:
AisC-R orN,
DisC-R4orN,
EisC-R6orNand
GisC-R7orN,
with the proviso that at least one of A, D, E and G is not N;
X, Y and Z are independently selected from the group consisting of: N and C-R8, with the proviso that at least one of X, Y and Z is not N;
Rl and R2 are each independently selected from the group consisting of: (1) hydogenand
(2) Ci_6alkyl, optionally substituted with 1 to 3 halo groups, or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form a 3- to 6-membered saturated monocyclic ring; R3, R4, R6 and R? are each independently selected from the group consisting of:
(1) hydrogen,
(2) halo (3) cyano, and
(4) Ci_4alkyl or Cι_4alkoxy, each optionally substituted with 1 to
3 halo groups;
R5 is selected from the group consisting of: (1) Ci-6alkyl,
(2) C2-6alkenyl,
(3) C2-6alkynyl,
(4) C3-6cycloalkyl,
(5) Cι_6alkoxy, (6) C3_6cycloalkoxy,
(7) Ci-6acyl,
(8) halo,
(9) aryl and
(10) HET, wherein groups (1) to (7) above are optionally substituted with from one up to the maximum number of substituable positions with halo, and
groups (9) and (10) above are optionally substituted with 1 to 3 substituents independently selected from the group consisting of: (a) halo, and
(b) C 1 -4alkyl or C i -4alkoxy, each optionally substituted with oxo, hydroxy or 1 to 3 halo groups,
or R4 and R5 may be joined together with the atoms to which they are attached to form a 5 or 6-membered monocyclic ring, optionally containing 1 to 3 heteratoms selected from O, S and NR8, said ring optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, Cι_4alkyl and Ci-4alkoxy, said Cι_4alkyl or Cι_4alkoxy optionally substituted with 1 to 3 halo groups; each R8 is independently selected from the group consisting of: hydrogen, halo and Ci-4alkyl, wherein said Cι_4alkyl is optionally substituted with 1 to 3 halo groups; and
HET is selected from the group consisting of: benzimidazolyl, benzofuranyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, imidazolyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridopyridinyl, pyridazinyl, pyridyl, pyrimidyl, pynolyl, quinazolinyl, quinolyl, quinoxalinyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, 1,4-dioxanyl, hexahydroazepinyl, piperazinyl, piperidinyl, pynolidinyl, morpholinyl, thiomorpholinyl, dihydrobenzimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropynolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, and tetrahydrothienyl.
2. The compound according to Claim 1 wherein:
A is N,
D is C-R4,
E is C-R6 and
G is C-R7.
The compound according to Claim 1 wherein: A is C-R3,
D is C-R4,
E is C-R6 and
G is C-R7.
4. The compound according to Claim 3 wherein X, Y and Z are
C-R8.
5. The compound according to Claim 3 wherein R3, R6 and R7 are hydrogen.
6. The compound according to Claim 5 wherein R4 is trifluoromethyl or cyano.
7. The compound according to Claim 3 wherein Rl and R are each independently selected from the group consisting of hydrogen, methyl and ethyl.
8. The compound according to Claim 3 wherein R5 is selected from the group consisting of:
(1) C2-6alkyl, (2) C3-6cycloalkyl,
(3) C2-6alkoxy,
(4) C3_6cycloalkoxy, and
(5) C3-6acyl, wherein groups (1) to (5) above are optionally substituted with 1 to 5 fluoro groups.
9. The compound according to Claim 8 wherein R5 is C2- 6alkoxy, optionally substituted with 1 to 5 fluoro groups.
10. The compound according to Claim 3 wherein R5 is selected from the group consisting of:
(1) phenyl, optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, methyl, methoxy and hydroxymethyl,
(2) oxadiazolyl,
(3) oxazolyl,
(4) furanyl and (5) thienyl.
11. The compound according to Claim 3 wherein X is N and Y and Z are both C-R8.
12. The compound according to Claim 3 wherein X and Z are both
C-R8 and Y is N.
13. The compound according to Claim 3, wherein:
Rl and R2 are each independently selected from the group consisting of: hydrogen and methyl,
R3, R6 and R7 are hydrogen,
R4 is trifluoromethyl or cyano, and
R5 is C2-6alkoxy, optionally substituted with 1 to 5 fluoro groups.
14. The compound according to Claim 13 wherein R5 is selected from 2,2,2-trifluoroethoxy and 2,2,2-trifluoro-l-methylethoxy.
15. The compound according to Claim 14 wherein X, Y and Z are C-R8 and each R8 is independently selected from hydrogen, methyl and halo.
-I ll-
16. The compound according to Claim 14 wherein X is N and Y and Z are both C-R8 and each R8 is independently selected from hydrogen, methyl and halo.
17. The compound according to Claim 14 wherein X and Z are both C-R8 and Y is N and each R8 is independently selected from hydrogen, methyl and halo.
18. A compound selected from the following table:
Figure imgf000113_0001
Figure imgf000114_0001
-113-
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
or a pharmaceutically acceptable salt of any of the above.
19. A method of treating an immunoregulatory abnormality in a mammalian patient in need of such treatment comprising administering to said patient a compound in accordance with Claim 1 in an amount that is effective for treating said immunoregulatory abnormality.
20. The method according to Claim 19 wherein the immunoregulatory abnormality is an autoimmune or chronic inflammatory disease selected from the group consisting of: systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cinhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy and asthma.
21. The method according to Claim 19 wherein the immunoregulatory abnormality is bone manow or organ transplant rejection or graft- versus-host disease.
22. The method according to Claim 19 wherein the immunoregulatory abnormality is selected from the group consisting of: transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, post-infectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, sebonhoeic dermatitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa, urticaria, angioedemas, vasculitis, erythema, cutaneous eosinophilia, lupus erythematosus, acne, alopecia areata, keratocoηjunctivitis, vernal conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis corneae, corneal leukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' opthalmopathy, Nogt-Koyanagi-Harada syndrome, sarcoidosis, pollen allergies, reversible obstructive airway disease, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, chronic or inveterate asthma, late asthma and airway hyper-responsiveness, bronchitis, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic bowel diseases, inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal burns, coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, ulcerative colitis, migraine, rhinitis, eczema, interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome, diabetic nephropathy, multiple myositis, Guillain-Bane syndrome, Meniere's disease, polyneuritis, multiple neuritis, mononeuritis, radiculopathy, hyperthyroidism, Basedow's disease, pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia, anerythroplasia, osteoporosis, sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photoallergic sensitivity, cutaneous T cell lymphoma, arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, myocardosis, scleroderma, Wegener's granuloma, Sjogren's syndrome, adiposis, eosinophilic fascitis, lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis, glomerulonephritis, male pattern alopecia or alopecia senilis by preventing epilation or providing hair germination and/or promoting hair generation and hair growth, muscular dystrophy, pyoderma and Sezary's syndrome, Addison's disease, ischemia-reperfusion injury of organs which occurs upon preservation, transplantation or ischemic disease, endotoxin-shock, pseudomembranous colitis, colitis caused by drug or radiation, ischemic acute renal insufficiency, chronic renal insufficiency, toxinosis caused by lung-oxygen or drugs, lung cancer, pulmonary emphysema, cataracta, siderosis, retinitis pigmentosa, senile macular degeneration, vitreal scaning, comeal alkali bum, dermatitis erythema multiforme, linear IgA ballous dermatitis and cement dermatitis, gingivitis, periodontitis, sepsis, pancreatitis, diseases caused by environmental pollution, aging, carcinogenesis, metastasis of carcinoma and hypobaropathy, disease caused by histamine or leukotriene-C4 release, Behcet's disease, autoimmune hepatitis, primary biliary cinhosis, sclerosing cholangitis, partial liver resection, acute liver necrosis, necrosis caused by toxin, viral hepatitis, shock, or anoxia, B-virus hepatitis, non- A/non-B hepatitis, cinhosis, alcoholic cinhosis, hepatic failure, fulminant hepatic failure, late-onset hepatic failure, "acute-on-chronic" liver failure, augmentation of chemotherapeutic effect, cytomegalovirus infection, HCMN infection, AIDS, cancer, senile dementia, trauma, and chronic bacterial infection.
23. The method according to Claim 19 wherein the immunoregulatory abnormality is selected from the group consisting of:
1) multiple sclerosis,
2) rheumatoid arthritis,
3) systemic lupus erythematosus, 4) psoriasis,
5) rej ection of transplanted organ or tissue,
6) inflammatory bowel disease,
7) a malignancy of lymphoid origin, 8) acute and chronic lymphocytic leukemias and lymphomas and
9) insulin and non-insulin dependent diabetes.
24. A method of suppressing the immune system in a mammalian patient in need of immunosuppression comprising administering to said patient an immunosuppressmg effective amount of a compound of Claim 1.
25. A pharmaceutical composition comprised of a compound in accordance with Claim 1 in combination with a pharmaceutically acceptable canier.
26. A method of treating a respiratory disease or condition in a mammalian patient in need of such treatment comprising administering to said patient a compound in accordance with Claim 1 in an amount that is effective for treating said respiratory disease or condition.
27. The method according to Claim 26 wherein the respiratory disease or condition is selected from the group consisting of: asthma, chronic bronchitis, chronic obstructive pulmonary disease, adult respiratory distress syndrome, infant respiratory distress syndrome, cough, eosinophilic granuloma, respiratory syncytial virus bronchiolitis, bronchiectasis, idiopathic pulmonary fibrosis, acute lung injury and bronchiolitis obliterans organizing pneumonia.
PCT/US2004/014837 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists WO2004103279A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2006532984A JP2006528980A (en) 2003-05-15 2004-05-12 3- (2-Amino-1-azacyclo) -5-aryl-1,2,4-oxadiazoles as S1P receptor agonists
EP04751981A EP1625123A4 (en) 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
CA002524867A CA2524867A1 (en) 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
AU2004240586A AU2004240586A1 (en) 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as S1P receptor agonists
US10/554,665 US20060252741A1 (en) 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47065903P 2003-05-15 2003-05-15
US60/470,659 2003-05-15

Publications (2)

Publication Number Publication Date
WO2004103279A2 true WO2004103279A2 (en) 2004-12-02
WO2004103279A3 WO2004103279A3 (en) 2005-05-19

Family

ID=33476733

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/014837 WO2004103279A2 (en) 2003-05-15 2004-05-12 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists

Country Status (7)

Country Link
US (1) US20060252741A1 (en)
EP (1) EP1625123A4 (en)
JP (1) JP2006528980A (en)
CN (1) CN1788008A (en)
AU (1) AU2004240586A1 (en)
CA (1) CA2524867A1 (en)
WO (1) WO2004103279A2 (en)

Cited By (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005058848A1 (en) * 2003-12-17 2005-06-30 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as s1p (edg) receptor agonists
WO2006088944A1 (en) * 2005-02-14 2006-08-24 University Of Virginia Patent Foundation Sphingosine 1- phos phate agonists comprising cycloalkanes and 5 -membered heterocycles substituted by amino and phenyl groups
WO2006100633A1 (en) * 2005-03-23 2006-09-28 Actelion Pharmaceuticals Ltd NOVEL THIOPHENE DERIVATIVES AS SPHINGOSINE-l-PHOSPHATE-1 RECEPTOR AGONISTS
US7241790B2 (en) 2002-07-30 2007-07-10 University Of Virginia Patent Foundation Compounds active in spinigosine 1-phosphate signaling
US7241812B2 (en) 2004-08-13 2007-07-10 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (S1P) receptor activity
WO2007085451A2 (en) * 2006-01-27 2007-08-02 Novartis Ag 3,5-di (aryl or heteroaryl) isoxazoles and 1, 2, 4-oxadiazoles as s1p1 receptor agonists, immunosuppresssive and anti -inflammatory agents
WO2007089018A1 (en) 2006-02-03 2007-08-09 Taisho Pharmaceutical Co., Ltd. Triazole derivative
WO2007092190A2 (en) * 2006-02-06 2007-08-16 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (s1p) receptor activity
WO2007091570A1 (en) 2006-02-06 2007-08-16 Taisho Pharmaceutical Co., Ltd. Binding inhibitor of sphingosine-1-phosphate
WO2007098474A1 (en) * 2006-02-21 2007-08-30 University Of Virginia Patent Foundation Phenyl-cycloalkyl and phenyl-heterocyclic derivatives as s1p receptor agonists
WO2007116866A1 (en) 2006-04-03 2007-10-18 Astellas Pharma Inc. Hetero compound
WO2008018447A1 (en) 2006-08-08 2008-02-14 Kyorin Pharmaceutical Co., Ltd. Aminoalcohol derivative and immunosuppressant containing the same as active ingredient
WO2008018427A1 (en) 2006-08-08 2008-02-14 Kyorin Pharmaceutical Co., Ltd. Aminophosphoric acid ester derivative and s1p receptor modulator containing the same as active ingredient
WO2008074820A1 (en) * 2006-12-21 2008-06-26 Glaxo Group Limited Oxadiazole derivatives as s1p1 receptor agonists
WO2008074821A1 (en) 2006-12-21 2008-06-26 Glaxo Group Limited Indole derivatives as s1p1 receptor agonists
WO2008114157A1 (en) * 2007-03-16 2008-09-25 Actelion Pharmaceuticals Ltd Amino- pyridine derivatives as s1p1 /edg1 receptor agonists
WO2009017219A1 (en) 2007-08-01 2009-02-05 Taisho Pharmaceutical Co., Ltd. Inhibitor of binding of s1p1
WO2009024905A1 (en) * 2007-08-17 2009-02-26 Actelion Pharmaceuticals Ltd Pyridine derivatives as s1p1/edg1 receptor modulators
WO2009057079A2 (en) * 2007-11-01 2009-05-07 Actelion Pharmaceuticals Ltd Novel pyrimidine derivatives
WO2009151621A1 (en) * 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
WO2009151626A1 (en) * 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
US7638637B2 (en) 2003-11-03 2009-12-29 University Of Virginia Patent Foundation Orally available sphingosine 1-phosphate receptor agonists and antagonists
JP2010504320A (en) * 2006-09-21 2010-02-12 アクテリオン ファーマシューティカルズ リミテッド Phenyl derivatives and their use as immunomodulators
US7723378B2 (en) 2005-03-23 2010-05-25 Actelion Pharmaceuticals Ltd. Hydrogenated benzo (C) thiophene derivatives as immunomodulators
US7750040B2 (en) 2004-07-29 2010-07-06 Actelion Pharmaceuticals Ltd Thiophene derivatives
WO2010081692A1 (en) * 2009-01-19 2010-07-22 Almirall, S.A. Oxadiazole derivatives as slpl receptor agonists
US7786173B2 (en) 2006-11-21 2010-08-31 University Of Virginia Patent Foundation Tetralin analogs having sphingosine 1-phosphate agonist activity
WO2010100142A1 (en) 2009-03-03 2010-09-10 Merck Serono S.A. Oxazole pyridine derivatives useful as s1p1 receptor agonists
US7915315B2 (en) 2006-11-21 2011-03-29 University Of Virginia Patent Foundation Benzocycloheptyl analogs having sphingosine 1-phosphate receptor activity
US7964649B2 (en) 2006-11-21 2011-06-21 University Of Virginia Patent Foundation Hydrindane analogs having sphingosine 1-phosphate receptor agonist activity
US8003800B2 (en) 2006-01-11 2011-08-23 Actelion Pharmaceuticals Ltd. Thiophene derivatives as S1P1/EDG1 receptor agonists
US8008286B2 (en) 2006-01-27 2011-08-30 University Of Virginia Patent Foundation Method for treatment of neuropathic pain
US8017631B2 (en) 2005-04-26 2011-09-13 Neurosearch A/S Oxadiazole derivatives and their medical use
US8022225B2 (en) 2004-08-04 2011-09-20 Taisho Pharmaceutical Co., Ltd Triazole derivative
US8133910B2 (en) 2006-09-07 2012-03-13 Actelion Pharmaceuticals Ltd. Thiophene derivatives as S1P1/EDGE1 receptor agonists
US8173710B2 (en) 2006-02-09 2012-05-08 University Of Virginia Patent Foundation Bicyclic sphingosine 1-phosphate analogs
US8178562B2 (en) 2006-01-24 2012-05-15 Actelion Pharmaceuticals, Ltd. Pyridine derivatives
US8188279B2 (en) 2008-05-08 2012-05-29 Allergan, Inc. Therapeutically useful substituted hydropyrido [3,2,1-ij] quinoline compounds
WO2012074719A1 (en) * 2010-12-03 2012-06-07 Allergan, Inc. Novel pyridine derivatives as sphingosine 1-phosphate (s1p) receptor modulators
WO2012080641A1 (en) * 2010-12-13 2012-06-21 Centre National De La Recherche Scientifique - Cnrs - S1p receptor agonists and use thereof in treating hiv infections
US8288554B2 (en) 2006-09-08 2012-10-16 Actelion Pharmaceuticals Ltd. Pyridin-3-yl derivatives as immunomodulating agents
US8329730B2 (en) 2008-04-30 2012-12-11 Glaxo Group Limited Compounds
US8399451B2 (en) 2009-08-07 2013-03-19 Bristol-Myers Squibb Company Heterocyclic compounds
US8410112B2 (en) 2008-11-10 2013-04-02 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8580824B2 (en) 2006-09-07 2013-11-12 Actelion Pharmaceuticals Ltd. Pyridin-4-yl derivatives as immunomodulating agents
US8580841B2 (en) 2008-07-23 2013-11-12 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8658675B2 (en) 2009-07-16 2014-02-25 Actelion Pharmaceuticals Ltd. Pyridin-4-yl derivatives
US8729062B2 (en) 2010-12-03 2014-05-20 Allergan, Inc. Benzyl azetidine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8735433B1 (en) 2012-11-14 2014-05-27 Allergan, Inc. Aryl oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US8835470B2 (en) 2010-04-23 2014-09-16 Bristol-Myers Squibb Company Mandelamide heterocyclic compounds
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841308B2 (en) 2008-12-19 2014-09-23 Vertex Pharmaceuticals Incorporated Pyrazin-2-amines useful as inhibitors of ATR kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846728B2 (en) 2010-12-03 2014-09-30 Allergan, Inc. Oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8871755B2 (en) 2013-02-12 2014-10-28 Allergan, Inc. Alkene azetidine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2015025001A (en) * 2007-10-04 2015-02-05 メルク セローノ ソシエテ アノニム Oxadiazole diaryl compounds
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
AU2013201157B2 (en) * 2006-12-21 2015-06-11 Glaxo Group Limited Indole derivatives as s1p1 receptor agonists
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US9096584B2 (en) 2010-05-12 2015-08-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
US9133179B2 (en) 2011-01-19 2015-09-15 Actelion Pharmaceuticals Ltd. 2-methoxy-pyridin-4-yl-derivatives
US9187437B2 (en) 2010-09-24 2015-11-17 Bristol-Myers Squibb Company Substituted oxadiazole compounds
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9630956B2 (en) 2010-05-12 2017-04-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US10385043B2 (en) 2015-05-20 2019-08-20 Idorsia Pharmaceuticals Ltd Crystalline form of the compound (S)-3-{4-[5-(2-cyclopentyl-6-methoxy-pyridin-4-yl)-[1,2,4]oxadiazol-3-yl]-2-ethyl-6-methyl-phenoxy}-propane-1,2-diol
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL1772145T3 (en) * 2004-07-16 2011-08-31 Kyorin Seiyaku Kk Method of effectively using medicine and method concerning prevention of side effect
KR101181090B1 (en) * 2004-10-12 2012-09-07 교린 세이야꾸 가부시키 가이샤 Process for producing 2-amino-2-[2-[4-3-benzyloxyphenylthio-2-chlorophenyl]ethyl]-1,3-propanediol hydrochloride or hydrate thereof and intermediate for the same
AU2006256968A1 (en) * 2005-06-08 2006-12-14 Novartis Ag Polycyclic oxadiazoles or I soxazoles and their use as SIP receptor ligands
PT1932522E (en) * 2005-10-07 2012-06-26 Kyorin Seiyaku Kk Therapeutic agent for liver disease containing 2-amino-1,3-propanediol derivative as active ingredient
TWI389683B (en) * 2006-02-06 2013-03-21 Kyorin Seiyaku Kk A therapeutic agent for an inflammatory bowel disease or an inflammatory bowel disease treatment using a 2-amino-1,3-propanediol derivative as an active ingredient
GB0625647D0 (en) * 2006-12-21 2007-01-31 Glaxo Group Ltd Compounds
US8383852B2 (en) * 2007-02-16 2013-02-26 Emisphere Technologies, Inc. Compounds having a cyclic moiety and compositions for delivering active agents
KR20090130062A (en) * 2007-04-19 2009-12-17 글락소 그룹 리미티드 Oxadiazole substituted indazole derivatives for use as sphingosine 1-phosphate (s1p) agonists
AR067762A1 (en) * 2007-07-31 2009-10-21 Vertex Pharma PROCESS TO PREPARE 5-FLUORO-1H-PIRAZOLO (3,4-B) PIRIDIN-3-AMINA AND DERIVATIVES OF THE SAME
AU2008306885B2 (en) * 2007-10-04 2013-12-05 Merck Serono S.A. Oxadiazole derivatives
TW200946105A (en) 2008-02-07 2009-11-16 Kyorin Seiyaku Kk Therapeutic agent or preventive agent for inflammatory bowel disease containing amino alcohol derivative as active ingredient
ES2389042T3 (en) * 2008-03-06 2012-10-22 Actelion Pharmaceuticals Ltd. Pyridine compounds
JP2012515789A (en) * 2009-01-23 2012-07-12 ブリストル−マイヤーズ スクイブ カンパニー Pyrazole-1,2,4-oxadiazole derivatives as sphingosine-1-phosphate agonists
US8354398B2 (en) * 2009-01-23 2013-01-15 Bristol-Myers Squibb Company Substituted isoxazole compounds
US8247436B2 (en) 2010-03-19 2012-08-21 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
WO2012061459A1 (en) * 2010-11-03 2012-05-10 Bristol-Myers Squibb Company Heterocyclic compounds as s1p1 agonists for the treatment of autoimmune and vascular diseases
CN108178759B (en) * 2018-01-05 2020-06-09 上海瑞纷医药科技有限责任公司 Synthesis method of α -adrenoceptor antagonist
WO2020051378A1 (en) 2018-09-06 2020-03-12 Arena Pharmaceuticals, Inc. Compounds useful in the treatment of autoimmune and inflammatory disorders

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3647809A (en) * 1968-04-26 1972-03-07 Chinoin Gyogyszer Es Vegyeszet Certain pyridyl-1 2 4-oxadiazole derivatives
DK1210344T3 (en) * 1999-08-19 2006-03-06 Nps Pharma Inc Heteropolycyclic Compounds and Their Uses as Metabotropic Glutamate Receptor Antagonists
NZ527691A (en) * 2001-02-21 2007-01-26 Nps Pharma Inc Heteropolycyclic compounds and their use as metabotropic glutamate receptor antagonists
US7479504B2 (en) * 2002-01-18 2009-01-20 Merck & Co., Inc. Edg receptor agonists
CA2488117A1 (en) * 2002-06-17 2003-12-24 Merck & Co., Inc. 1-((5-aryl-1,2,4-oxadiazol-3-yl)benzyl)azetidine-3-carboxylates and 1-((5-aryl-1,2,4-oxadiazol-3-yl)benzyl)pyrrolidine-3-carboxylates as edg receptor agonists

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1625123A4 *

Cited By (173)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7560477B2 (en) 2002-07-30 2009-07-14 University Of Virginia Patent Foundation Compounds active in sphingosine 1-phosphate signaling
US7241790B2 (en) 2002-07-30 2007-07-10 University Of Virginia Patent Foundation Compounds active in spinigosine 1-phosphate signaling
US7638637B2 (en) 2003-11-03 2009-12-29 University Of Virginia Patent Foundation Orally available sphingosine 1-phosphate receptor agonists and antagonists
US7605171B2 (en) 2003-12-17 2009-10-20 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as S1P (Edg) receptor agonists
WO2005058848A1 (en) * 2003-12-17 2005-06-30 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as s1p (edg) receptor agonists
AU2004299456B2 (en) * 2003-12-17 2010-10-07 Merck Sharp & Dohme Corp. (3,4-disubstituted)propanoic carboxylates as S1P (Edg) receptor agonists
US7750040B2 (en) 2004-07-29 2010-07-06 Actelion Pharmaceuticals Ltd Thiophene derivatives
US8022225B2 (en) 2004-08-04 2011-09-20 Taisho Pharmaceutical Co., Ltd Triazole derivative
US7241812B2 (en) 2004-08-13 2007-07-10 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (S1P) receptor activity
WO2006088944A1 (en) * 2005-02-14 2006-08-24 University Of Virginia Patent Foundation Sphingosine 1- phos phate agonists comprising cycloalkanes and 5 -membered heterocycles substituted by amino and phenyl groups
US7754703B2 (en) 2005-02-14 2010-07-13 University Of Virginia Patent Foundation Cycloalkane-containing sphingosine 1-phosphate agonists
US8329676B2 (en) 2005-02-14 2012-12-11 University Of Virginia Patent Foundation Cycloalkane-containing sphingosine 1-phosphate agonists
JP2008530135A (en) * 2005-02-14 2008-08-07 ユニバーシティ オブ バージニア パテント ファンデーション Sphingosine = 1-phosphate agonist containing a cycloalkane substituted with an amino group and a phenyl group and a 5-membered heterocyclic ring
US8039644B2 (en) 2005-03-23 2011-10-18 Actelion Pharmaceuticals Ltd. Hydrogenated benzo (C) thiophene derivatives as immunomodulators
US7723378B2 (en) 2005-03-23 2010-05-25 Actelion Pharmaceuticals Ltd. Hydrogenated benzo (C) thiophene derivatives as immunomodulators
US7605269B2 (en) 2005-03-23 2009-10-20 Actelion Pharmaceuticals Ltd. Thiophene derivatives as Sphingosine-1-phosphate-1 receptor agonists
WO2006100633A1 (en) * 2005-03-23 2006-09-28 Actelion Pharmaceuticals Ltd NOVEL THIOPHENE DERIVATIVES AS SPHINGOSINE-l-PHOSPHATE-1 RECEPTOR AGONISTS
US8017631B2 (en) 2005-04-26 2011-09-13 Neurosearch A/S Oxadiazole derivatives and their medical use
US8003800B2 (en) 2006-01-11 2011-08-23 Actelion Pharmaceuticals Ltd. Thiophene derivatives as S1P1/EDG1 receptor agonists
US8178562B2 (en) 2006-01-24 2012-05-15 Actelion Pharmaceuticals, Ltd. Pyridine derivatives
US8697732B2 (en) 2006-01-24 2014-04-15 Actelion Pharmaceuticals Ltd. Pyridine derivatives
WO2007085451A3 (en) * 2006-01-27 2007-12-21 Novartis Ag 3,5-di (aryl or heteroaryl) isoxazoles and 1, 2, 4-oxadiazoles as s1p1 receptor agonists, immunosuppresssive and anti -inflammatory agents
JP2009524611A (en) * 2006-01-27 2009-07-02 ノバルティス アクチエンゲゼルシャフト Reverse isoxazole
US7799812B2 (en) 2006-01-27 2010-09-21 Novartis Ag Reverse isoxazoles
US8008286B2 (en) 2006-01-27 2011-08-30 University Of Virginia Patent Foundation Method for treatment of neuropathic pain
WO2007085451A2 (en) * 2006-01-27 2007-08-02 Novartis Ag 3,5-di (aryl or heteroaryl) isoxazoles and 1, 2, 4-oxadiazoles as s1p1 receptor agonists, immunosuppresssive and anti -inflammatory agents
JP5035752B2 (en) * 2006-02-03 2012-09-26 大正製薬株式会社 Triazole derivative
WO2007089018A1 (en) 2006-02-03 2007-08-09 Taisho Pharmaceutical Co., Ltd. Triazole derivative
US8022091B2 (en) 2006-02-03 2011-09-20 Taisho Pharmaceutical Co., Ltd. Triazole derivative
WO2007091570A1 (en) 2006-02-06 2007-08-16 Taisho Pharmaceutical Co., Ltd. Binding inhibitor of sphingosine-1-phosphate
WO2007092190A3 (en) * 2006-02-06 2007-11-29 Praecis Pharm Inc Methods and compositions for modulating sphingosine-1-phosphate (s1p) receptor activity
US7994204B2 (en) 2006-02-06 2011-08-09 Taisho Pharmaceutical Co., Ltd Binding inhibitor of sphingosine-1-phosphate
WO2007092190A2 (en) * 2006-02-06 2007-08-16 Praecis Pharmaceuticals, Inc. Methods and compositions for modulating sphingosine-1-phosphate (s1p) receptor activity
US8173710B2 (en) 2006-02-09 2012-05-08 University Of Virginia Patent Foundation Bicyclic sphingosine 1-phosphate analogs
WO2007098474A1 (en) * 2006-02-21 2007-08-30 University Of Virginia Patent Foundation Phenyl-cycloalkyl and phenyl-heterocyclic derivatives as s1p receptor agonists
WO2007116866A1 (en) 2006-04-03 2007-10-18 Astellas Pharma Inc. Hetero compound
US7678820B2 (en) 2006-04-03 2010-03-16 Astellas Pharma Inc. Hetero compound
US7951825B2 (en) 2006-04-03 2011-05-31 Astellas Pharma Inc. Hetero compound
WO2008018427A1 (en) 2006-08-08 2008-02-14 Kyorin Pharmaceutical Co., Ltd. Aminophosphoric acid ester derivative and s1p receptor modulator containing the same as active ingredient
WO2008018447A1 (en) 2006-08-08 2008-02-14 Kyorin Pharmaceutical Co., Ltd. Aminoalcohol derivative and immunosuppressant containing the same as active ingredient
US8133910B2 (en) 2006-09-07 2012-03-13 Actelion Pharmaceuticals Ltd. Thiophene derivatives as S1P1/EDGE1 receptor agonists
US8580824B2 (en) 2006-09-07 2013-11-12 Actelion Pharmaceuticals Ltd. Pyridin-4-yl derivatives as immunomodulating agents
US8288554B2 (en) 2006-09-08 2012-10-16 Actelion Pharmaceuticals Ltd. Pyridin-3-yl derivatives as immunomodulating agents
US8044076B2 (en) 2006-09-21 2011-10-25 Actelion Pharmaceuticals Ltd. Phenyl derivatives and their use as immunomodulators
JP2010504320A (en) * 2006-09-21 2010-02-12 アクテリオン ファーマシューティカルズ リミテッド Phenyl derivatives and their use as immunomodulators
US7915315B2 (en) 2006-11-21 2011-03-29 University Of Virginia Patent Foundation Benzocycloheptyl analogs having sphingosine 1-phosphate receptor activity
US7964649B2 (en) 2006-11-21 2011-06-21 University Of Virginia Patent Foundation Hydrindane analogs having sphingosine 1-phosphate receptor agonist activity
US7786173B2 (en) 2006-11-21 2010-08-31 University Of Virginia Patent Foundation Tetralin analogs having sphingosine 1-phosphate agonist activity
AU2013201157B2 (en) * 2006-12-21 2015-06-11 Glaxo Group Limited Indole derivatives as s1p1 receptor agonists
WO2008074820A1 (en) * 2006-12-21 2008-06-26 Glaxo Group Limited Oxadiazole derivatives as s1p1 receptor agonists
US8101775B2 (en) 2006-12-21 2012-01-24 Glaxo Group Limited Indole derivatives as S1P1 Receptor
JP2010513397A (en) * 2006-12-21 2010-04-30 グラクソ グループ リミテッド Indole derivatives as S1P1 receptor agonists
EA017406B1 (en) * 2006-12-21 2012-12-28 Глэксо Груп Лимитед Indole derivatives as s1p1 receptor agonists
WO2008074821A1 (en) 2006-12-21 2008-06-26 Glaxo Group Limited Indole derivatives as s1p1 receptor agonists
EP2206710A1 (en) * 2006-12-21 2010-07-14 Glaxo Group Limited Indole derivatives as S1P1 receptor agonists
WO2008114157A1 (en) * 2007-03-16 2008-09-25 Actelion Pharmaceuticals Ltd Amino- pyridine derivatives as s1p1 /edg1 receptor agonists
AU2008227979B2 (en) * 2007-03-16 2014-02-06 Actelion Pharmaceuticals Ltd Amino- pyridine derivatives as S1P1 /EDG1 receptor agonists
US8592460B2 (en) 2007-03-16 2013-11-26 Actelion Pharmaceuticals Ltd. Amino-pyridine derivatives as S1P1 /EDG1 receptor agonists
KR101454944B1 (en) * 2007-03-16 2014-10-27 액테리온 파마슈티칼 리미티드 Amino-pyridine derivatives as s1p1/edg1 receptor agonists
US8048898B2 (en) 2007-08-01 2011-11-01 Taisho Pharmaceutical Co., Ltd Inhibitor of binding of S1P1
WO2009017219A1 (en) 2007-08-01 2009-02-05 Taisho Pharmaceutical Co., Ltd. Inhibitor of binding of s1p1
US8598208B2 (en) 2007-08-17 2013-12-03 Actelion Pharmaceuticals Ltd. Pyridine derivatives as S1P1/EDG1 receptor modulators
WO2009024905A1 (en) * 2007-08-17 2009-02-26 Actelion Pharmaceuticals Ltd Pyridine derivatives as s1p1/edg1 receptor modulators
JP2015025001A (en) * 2007-10-04 2015-02-05 メルク セローノ ソシエテ アノニム Oxadiazole diaryl compounds
WO2009057079A2 (en) * 2007-11-01 2009-05-07 Actelion Pharmaceuticals Ltd Novel pyrimidine derivatives
US8299086B2 (en) 2007-11-01 2012-10-30 Actelion Pharmaceuticals Ltd. Pyrimidine derivatives
WO2009057079A3 (en) * 2007-11-01 2009-12-03 Actelion Pharmaceuticals Ltd Novel pyrimidine derivatives
US8329730B2 (en) 2008-04-30 2012-12-11 Glaxo Group Limited Compounds
US8431702B2 (en) 2008-05-08 2013-04-30 Allergan, Inc. Therapeutically useful substituted hydropyrido [3,2,1-ij] quinoline compounds
US8309729B2 (en) 2008-05-08 2012-11-13 Allergan, Inc. Therapeutically useful substituted hydropyrido [3,2,1-ij] quinoline compounds
US8188279B2 (en) 2008-05-08 2012-05-29 Allergan, Inc. Therapeutically useful substituted hydropyrido [3,2,1-ij] quinoline compounds
WO2009151626A1 (en) * 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
WO2009151621A1 (en) * 2008-06-13 2009-12-17 Arena Pharmaceuticals, Inc. Substituted (1, 2, 4-0xadiaz0l-3-yl) indolin-1-yl carboxylic acid derivatives useful as s1p1 agonists
US9522133B2 (en) 2008-07-23 2016-12-20 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US8580841B2 (en) 2008-07-23 2013-11-12 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9126932B2 (en) 2008-07-23 2015-09-08 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US8415484B2 (en) 2008-08-27 2013-04-09 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US9108969B2 (en) 2008-08-27 2015-08-18 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US8410112B2 (en) 2008-11-10 2013-04-02 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841308B2 (en) 2008-12-19 2014-09-23 Vertex Pharmaceuticals Incorporated Pyrazin-2-amines useful as inhibitors of ATR kinase
US9365557B2 (en) 2008-12-19 2016-06-14 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US10479784B2 (en) 2008-12-19 2019-11-19 Vertex Pharmaceuticals Incorporated Substituted pyrazin-2-amines as inhibitors of ATR kinase
US9701674B2 (en) 2008-12-19 2017-07-11 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
US10961232B2 (en) 2008-12-19 2021-03-30 Vertex Pharmaceuticals Incorporated Substituted pyrazines as ATR kinase inhibitors
EP2210890A1 (en) * 2009-01-19 2010-07-28 Almirall, S.A. Oxadiazole derivatives as S1P1 receptor agonists
WO2010081692A1 (en) * 2009-01-19 2010-07-22 Almirall, S.A. Oxadiazole derivatives as slpl receptor agonists
AU2010220338B2 (en) * 2009-03-03 2016-07-21 Merck Serono S.A. Oxazole pyridine derivatives useful as S1P1 receptor agonists
US8791142B2 (en) 2009-03-03 2014-07-29 Merck Serono S.A. Oxazole pyridine derivatives useful as S1P1 receptor agonists
WO2010100142A1 (en) 2009-03-03 2010-09-10 Merck Serono S.A. Oxazole pyridine derivatives useful as s1p1 receptor agonists
US8658675B2 (en) 2009-07-16 2014-02-25 Actelion Pharmaceuticals Ltd. Pyridin-4-yl derivatives
US8399451B2 (en) 2009-08-07 2013-03-19 Bristol-Myers Squibb Company Heterocyclic compounds
US9175320B2 (en) 2010-01-27 2015-11-03 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9447041B2 (en) 2010-01-27 2016-09-20 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US8853419B2 (en) 2010-01-27 2014-10-07 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US11674163B2 (en) 2010-01-27 2023-06-13 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US11149292B2 (en) 2010-01-27 2021-10-19 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US8835470B2 (en) 2010-04-23 2014-09-16 Bristol-Myers Squibb Company Mandelamide heterocyclic compounds
US9062008B2 (en) 2010-05-12 2015-06-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8969356B2 (en) 2010-05-12 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9630956B2 (en) 2010-05-12 2017-04-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8962631B2 (en) 2010-05-12 2015-02-24 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9096584B2 (en) 2010-05-12 2015-08-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9334244B2 (en) 2010-05-12 2016-05-10 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8623869B2 (en) 2010-06-23 2014-01-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9187437B2 (en) 2010-09-24 2015-11-17 Bristol-Myers Squibb Company Substituted oxadiazole compounds
WO2012074898A1 (en) * 2010-12-03 2012-06-07 Allergan, Inc. Novel alkene derivatives as sphingosine 1-phosphate (s1p) receptor modulators
CN103338769A (en) * 2010-12-03 2013-10-02 阿勒根公司 Novel alkene derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8846728B2 (en) 2010-12-03 2014-09-30 Allergan, Inc. Oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8697733B2 (en) 2010-12-03 2014-04-15 Allergan, Inc. Pyridine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8288555B2 (en) 2010-12-03 2012-10-16 Allergan, Inc. Alkene derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8399492B2 (en) 2010-12-03 2013-03-19 Allergan, Inc. Pyridine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
CN103338770A (en) * 2010-12-03 2013-10-02 阿勒根公司 Novel pyridine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
WO2012074718A1 (en) * 2010-12-03 2012-06-07 Allergan, Inc. Novel oxadiazole derivatives as sphingosine 1-phosphate (s1p) receptor modulators
US8906899B2 (en) 2010-12-03 2014-12-09 Allergan, Inc. Azetidine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
CN103370067A (en) * 2010-12-03 2013-10-23 阿勒根公司 Novel oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8492410B2 (en) 2010-12-03 2013-07-23 Allergan, Inc. Pyridine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
WO2012074719A1 (en) * 2010-12-03 2012-06-07 Allergan, Inc. Novel pyridine derivatives as sphingosine 1-phosphate (s1p) receptor modulators
US8729062B2 (en) 2010-12-03 2014-05-20 Allergan, Inc. Benzyl azetidine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US8618139B2 (en) 2010-12-03 2013-12-31 Allergan, Inc. Oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
WO2012080641A1 (en) * 2010-12-13 2012-06-21 Centre National De La Recherche Scientifique - Cnrs - S1p receptor agonists and use thereof in treating hiv infections
US9133179B2 (en) 2011-01-19 2015-09-15 Actelion Pharmaceuticals Ltd. 2-methoxy-pyridin-4-yl-derivatives
US8877759B2 (en) 2011-04-05 2014-11-04 Vertex Pharnaceuticals Incorporated Aminopyrazines as ATR kinase inhibitors
US9309250B2 (en) 2011-06-22 2016-04-12 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-b]pyrazines as ATR kinase inhibitors
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US9096602B2 (en) 2011-06-22 2015-08-04 Vertex Pharmaceuticals Incorporated Substituted pyrrolo[2,3-B]pyrazines as ATR kinase inhibitors
US10822331B2 (en) 2011-09-30 2020-11-03 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US9862709B2 (en) 2011-09-30 2018-01-09 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US8765751B2 (en) 2011-09-30 2014-07-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846686B2 (en) 2011-09-30 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10208027B2 (en) 2011-09-30 2019-02-19 Vertex Pharmaceuticals Incorporated Processes for preparing ATR inhibitors
US10813929B2 (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating cancer with ATR inhibitors
US9035053B2 (en) 2011-09-30 2015-05-19 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of ATR kinase
US8846917B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8846918B2 (en) 2011-11-09 2014-09-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841449B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8841337B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11110086B2 (en) 2012-04-05 2021-09-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US10478430B2 (en) 2012-04-05 2019-11-19 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase and combination therapies thereof
US9791456B2 (en) 2012-10-04 2017-10-17 Vertex Pharmaceuticals Incorporated Method for measuring ATR inhibition mediated increases in DNA damage
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8735433B1 (en) 2012-11-14 2014-05-27 Allergan, Inc. Aryl oxadiazole derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US11117900B2 (en) 2012-12-07 2021-09-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10392391B2 (en) 2012-12-07 2019-08-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10787452B2 (en) 2012-12-07 2020-09-29 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11370798B2 (en) 2012-12-07 2022-06-28 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9340546B2 (en) 2012-12-07 2016-05-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9718827B2 (en) 2012-12-07 2017-08-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9650381B2 (en) 2012-12-07 2017-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US8871755B2 (en) 2013-02-12 2014-10-28 Allergan, Inc. Alkene azetidine derivatives as sphingosine 1-phosphate (S1P) receptor modulators
US9663519B2 (en) 2013-03-15 2017-05-30 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10160760B2 (en) 2013-12-06 2018-12-25 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11485739B2 (en) 2013-12-06 2022-11-01 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10815239B2 (en) 2013-12-06 2020-10-27 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10093676B2 (en) 2014-06-05 2018-10-09 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US9670215B2 (en) 2014-06-05 2017-06-06 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US10800781B2 (en) 2014-06-05 2020-10-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
US11179394B2 (en) 2014-06-17 2021-11-23 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of Chk1 and ATR inhibitors
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11896578B2 (en) 2015-01-06 2024-02-13 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US10836754B2 (en) 2015-05-20 2020-11-17 Idorsia Pharmaceuticals Ltd Crystalline form of the compound (S)-3-{4-[5-(2-cyclopentyl-6-methoxy-pyridin-4-yl)-[1,2,4]oxadiazol-3-yl]-2-ethyl-6-methyl-phenoxy}-propane-1,2-diol
US10385043B2 (en) 2015-05-20 2019-08-20 Idorsia Pharmaceuticals Ltd Crystalline form of the compound (S)-3-{4-[5-(2-cyclopentyl-6-methoxy-pyridin-4-yl)-[1,2,4]oxadiazol-3-yl]-2-ethyl-6-methyl-phenoxy}-propane-1,2-diol
US11390615B2 (en) 2015-05-20 2022-07-19 Idorsia Pharmaceuticals Ltd Crystalline form of the compound (S)-3-{4-[5-(2-cyclopentyl-6-methoxy-pyridin-4-yl)-[1,2,4]oxadiazol-3-yl]-2-ethyl-6-methyl-phenox
US11834443B2 (en) 2015-05-20 2023-12-05 Idorsia Pharmaceuticals Ltd Crystalline form of the compound (s)-3-{4-[5-(2-cyclopentyl-6-methoxy-pyridin-4-yl)-[1,2,4]oxadiazol-3-yl]-2-ethyl-6-methyl-phenoxy}-propane-1,2-diol
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US10676435B2 (en) 2015-06-22 2020-06-09 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound 1) for use in SIPI receptor-associated disorders
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
US11091435B2 (en) 2015-06-22 2021-08-17 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3, 4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(compound1) for use in S1P1 receptor-associated disorders
US11464774B2 (en) 2015-09-30 2022-10-11 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA damaging agents and ATR inhibitors
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations

Also Published As

Publication number Publication date
CN1788008A (en) 2006-06-14
WO2004103279A3 (en) 2005-05-19
EP1625123A2 (en) 2006-02-15
JP2006528980A (en) 2006-12-28
US20060252741A1 (en) 2006-11-09
EP1625123A4 (en) 2007-08-29
CA2524867A1 (en) 2004-12-02
AU2004240586A1 (en) 2004-12-02

Similar Documents

Publication Publication Date Title
EP1625123A2 (en) 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
AU2003297232B2 (en) 1-(amino)indanes and (1,2-dihydro-3-amino)-benzofurans, benzothiophenes and indoles
US7199142B2 (en) 1-((5-aryl-1,2,4-oxadiazol-3-yl) benzyl)azetidine-3-carboxylates and 1-((5-aryl-1,2,4-oxadiazol-3-yl)benzyl) pyrrolidine-3-carboxylates as edg receptor agonists
EP1670463A2 (en) 3,5-aryl, heteroaryl or cycloalkyl substituted-1,2,4-oxadiazoles as s1p receptor agonists
JP4709488B2 (en) N- (benzyl) aminoalkylcarboxylic acid compounds, phosphinic acid compounds, phosphonic acid compounds and tetrazoles as Edg receptor agonists
US7479504B2 (en) Edg receptor agonists
JP2005531506A (en) Aminoalkylphosphonates and related compounds as agonists of EDG receptors
EP1804793A2 (en) 2-(aryl)azacyclylmethyl carboxylates, sulfonates, phosphonates, phosphinates and heterocycles as s1p receptor agonists
AU2003202994A1 (en) N-(benzyl)aminoalkylcarboxylates, phosphinates, phosphonates and tetrazoles as Edg receptor agonists
WO2003074008A2 (en) Aminoalkylphosphonates and related compounds as edg receptor agonists

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004240586

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006252741

Country of ref document: US

Ref document number: 10554665

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 4936/DELNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2524867

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2004240586

Country of ref document: AU

Date of ref document: 20040512

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004240586

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 20048129905

Country of ref document: CN

Ref document number: 2006532984

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2004751981

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004751981

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10554665

Country of ref document: US