WO2007070961A1 - Mif inhibitors - Google Patents

Mif inhibitors Download PDF

Info

Publication number
WO2007070961A1
WO2007070961A1 PCT/AU2006/001965 AU2006001965W WO2007070961A1 WO 2007070961 A1 WO2007070961 A1 WO 2007070961A1 AU 2006001965 W AU2006001965 W AU 2006001965W WO 2007070961 A1 WO2007070961 A1 WO 2007070961A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
independently selected
compound
disease
diseases
Prior art date
Application number
PCT/AU2006/001965
Other languages
French (fr)
Inventor
Eric Francis Morand
Colin Edward Skene
Peter Mark Tapley
Xinhua Li
Thomas H. Jozefiak
Original Assignee
Cortical Pty Ltd
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 Cortical Pty Ltd filed Critical Cortical Pty Ltd
Priority to US12/158,563 priority Critical patent/US20090130165A1/en
Priority to EP06840387A priority patent/EP1968576A4/en
Priority to AU2006326850A priority patent/AU2006326850A1/en
Priority to CA002634212A priority patent/CA2634212A1/en
Priority to JP2008546033A priority patent/JP2009521415A/en
Publication of WO2007070961A1 publication Critical patent/WO2007070961A1/en
Priority to IL192331A priority patent/IL192331A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F2/00Filters implantable into blood vessels; Prostheses, i.e. artificial substitutes or replacements for parts of the body; Appliances for connecting them with the body; Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • A61F2/82Devices providing patency to, or preventing collapsing of, tubular structures of the body, e.g. stents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • 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
    • 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
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/02Drugs for disorders of the endocrine system of the hypothalamic hormones, e.g. TRH, GnRH, CRH, GRH, somatostatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/58Benzoxazoles; Hydrogenated benzoxazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2

Definitions

  • the present invention relates generally to the treatment of diseases or conditions resulting from cellular activation, such as inflammatory or cancerous diseases or conditions.
  • the invention relates to the use of specific bcnzimidazolone analogues and derivatives to inhibit the cytokine or biological activity of macrophage migration inhibitory factor (MIF), and diseases or conditions wherein MIF cytokine or biological activity is implicated.
  • MIF macrophage migration inhibitory factor
  • MIF is the first identified T-cell-derived soluble lymphoki ⁇ e. MIF was first described as a soluble factor with the ability to modify the migration of macrophages ⁇ 1 K The molecule responsible for the biological actions ascribed to MIF was identified and 'cloned in 1989 (2 l Initially found to activate macrophages at inflammatoiy sites, it has been shown to possess plurip ⁇ tential actions in the immune system. MIF has been shown to be expressed in human diseases which include inflammation, injury, ischaeroia or malignancy. MIF also has a unique relationship with glucocorticoids by overriding their and- inflammatory effects.
  • Antibody antagonism of MIF has also been shown to have activity in adjuvant- or collagen-induced arthritis animal models and models of other inflammatory and immune diseases including colitis, multiple sclerosis, atherosclerosis, glomerulonephritis, and uveitis.
  • glucocorticoids have been used to treat human diseases for over fifty years and are effective in ⁇ range of diseases which include inflammation, injury, ischacmia or malignancy. Although debate continues in relation to their impact on disease progression, their influence on symptoms and signs of inflammation, especially in the short term, can be dramatic.
  • glucocorticoids is limited by universal, predictable, dose- dependent toxicity. Mimicking Cushing's disease, a disease wherein the adrenal glands produce excess endogenous glucocorticoids, glucocorticoid treatment is associated with side effects including immunosuppression (resulting in increased susceptibility to infections), weight gain, change in body habitus, hypertension, oedema, diabetes mellitus, cataracts, osteoporosis, poor wound healing, thinning of the skin, vascular fragility, hirsutism and other features of masculinization (in Females), In children, growth retardation is also noted. These side effects are known as Cushiiigoid side effects.
  • glucocorticoids are dose dependent, attempts to reduce the dosage requirement have been investigated, including combination therapies in which glucocorticoids are administered with other therapeutic agents. These combination therapies arc sometimes referred to as "steroid-sparing" therapies. However, currently available combination therapies are non-specific as the other therapeutic agents do not address biological events which inhibit the effectiveness of glucocorticoids. Such combination therapies are also typically associated with serious side effects.
  • glucocorticoids arc incompletely effective in a number of disease settings, leading to the concept of "steroid-resistant” diseases. Agents which amplify or enhance the effects of glucocorticoids would not only allow the reduction of dose of these agents but may also potentially fender "steroid-resistant” diseases steroid-sensitive.
  • Therapeutic antagonism of MTF may provide "steroid -sparing" effects or be therapeutic in "steroid-resistant” diseases.
  • the expression and/or release of M IF can be induced by glucocorticoids ⁇ ' ⁇ 4 ⁇
  • MlF is able to directly antagonize the effects of glucocorticoids. This has been shown to be the case for macrophage TNF, IL-I ⁇ , (L-6 and 1L-8 secretion (5X (6> , and for T cell proliferation and IL-2 release (7) .
  • MlF exerts a powerful glucocorticoid-atitagonist effect in models including e ⁇ dotoxic shock and experimental arthritis fs)l ⁇ K fn the context of an inflammatory or other disease treated with glucocorticoids, then, MIF is expressed but exerts an effect which prevents the glucocorticoid inhibition of inflammation. It can therefore be proposed that therapeutic antagonism of MIF would remove MIF' s role in inhibiting the anti-inflammatory effect of glucocorticoids, thereby allowing glucocorticoids to prevail. This would be the first example of true "steroid-sparing" therapy.
  • MIF has recently been shown to be important in the control of ieukocytc-cndothelial interactions.
  • Leukocytes interact with vascular endothelial cells in order to gain egress from the vasculature into tissues.
  • the role of MIF in this process has been demonstrated to affect in particular lcukocytc-endotlieliiil adhesion and emigration (22 ⁇ 23) .
  • This process is an essential part of nearly all inflammatory diseases, and also for diseases less well-identified as inflammatory including atherosclerosis (24) .
  • WO 03/104203 the present applicant has shown that certain benzimidazolc derivatives are capable of acting as inhibitors of MIF.
  • the present inventors have now found a novel class of MlF inhibitors, members of which show imp ⁇ oved characteristics as drug-like molecules when compared to the compounds of the prior art.
  • the present invention provides a method of treating, diagnosing or preventing autoimmune diseases, tumours, or chronic or acute inflammatory diseases comprising administering a treatment, prevention or diagnostic effective amount of a. compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof wherein:
  • X is selected from- -O-, -S-, -C(R 5 )(Ry)- and -N(R n )-;
  • Y is selected from - -N(R 7 )-, -O-, -S-, and -C(R 7 J 2 -;
  • R is selected from hydrogen, Ci-Q,alkyl, C(RsRsOi 1 SR 7 . (CRsRs 1 X 1 N(TIs) 2 and (CR 5 R 5 -X 4 TIaIo;
  • R 3 is selected from hydrogen, C,-C fi alkyl, (CR 16 R 16 O p NR 14 R is, (CR 1 ⁇ sR ⁇ v) r OR, 7 , (CR 16 R 1 6 OpSR 171 (CR
  • R 4 is selected from hydrogen, halogen, Ci-Csalkyl, C ⁇ -Caalkenyl, C 2 -C:salkynyl and
  • each Rs and R.y is independently selected from hydrogen, C]-Q>alkyl, halo, OR 7 , SR 7 and N(Rn) 2 ;
  • each R n is independently selected from hydrogen, Ci-Csalkyl and 0Ii 7 ;
  • each R 7 is independently selected from hydrogen and Ci-C3alkyl
  • each R 1 . 2 and R 12' is independently selected from hydrogen, Ci-Csalkyl, C2-C 6 alkenyl, C 2 - C ⁇ alkynyl, OR 24 , SR 24 , halo, N(R 24 )Z, CO 2 R 24 , CN, NO 2 , aryl and heteracyclyl;
  • each RM and Rn is independently selected from hydrogen, OR] 7 , SR 17 , and
  • each R 1 ri and R 1 ⁇ is independently selected from hydrogen, CrQalkyl, halo, OR ]7 , SR] 7 and N(R I7 ) 2 ;
  • each R )7 is independently selected from hydrogen and Ci-Csalkyl
  • each R 1 g is independently selected from hydrogen and halo
  • R 22 is selected from Ci-Qalkyl, NH 2 , NH(C ⁇ -C6alkyl), N(Ci-C c aIkyl) 2 , OR 29 or SR29;
  • each R 24 is selected from H and Ci-Cgalkyl;
  • R. 2 8 is selected from hydrogen, Ci-C 6 aLkyl, OR29, SR29 or N(R 2P ) 2 ;
  • each R 20 is independently selected from hydrogen and
  • Q is selected from O, S, NR 40 , S(O) 11 where u is an integer from ⁇ to 2;
  • R 4 Q is selected from H, OH, and C(R 4I R 1 UOvR 4 Z;
  • each R 41 and R 4 ⁇ is independently selected from H, OH, halo, NH 2 , cya ⁇ o, and NO 2 ;
  • R 42 is independently selected from H 1 OR 43 , COOR43, CON(R 43 ⁇ 1 ), O(CO)R43, aryl, and heterocyclyl;
  • each R 43 and Ro- is independently selected from H, Cj oalkyl, benzyl, and aryl;
  • a O or an integer to 3
  • n is 0 or an integer from 1 to 20;
  • p is 0 or an integer from 1 to 6;
  • I is an integer from 1 toll
  • v is 0 or an integer from i to 10.
  • the autoimmune disease, tumour, or chronic or acute inflammatory disease is selected from the group comprising:
  • rheumatic diseases including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis
  • spondyloarthropathies including but not limited to ankylosing spondylitis, reactive arthritis, Reiler's syndrome
  • crystal arthropathies including but not limited to gout, pscudogout, calcium pyrophosphate deposition disease
  • Lyme disease polymyalgia rhcumatica
  • connective tissue diseases including but not limited to systemic lupus syndrome
  • vasculitides including but not. limited to polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome;
  • vascular diseases including atlicrosclciOtic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischaemic heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis;
  • ocular diseases including uveitis, corneal disease, ulcerative colitis, cataracts; autoimmune diseases (including but not limited to diabetes mcllitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis);
  • pulmonary diseases including but not. limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome;
  • cancers whether primary or metastatic including but not. limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
  • renal diseases including glomerulonephritis, interstitial nephritis;
  • hypolhalamic-piluiiary-adrenal axis disorders of the hypolhalamic-piluiiary-adrenal axis
  • nervous system disorders including multiple sclerosis, Alzheimer's disease;
  • aiigiogenesis eg diabetic retinopathy, rheumatoid arthritis, cancer
  • endometrial function menstruation, implantation.
  • complications of infective disorders including endotoxic (septic) shock, cxotosic (septic) shock, infective (true septic) shock, malarial complications ⁇ other complications of infection, pelvic inflammatory disease;
  • transplant rejection graft-versus-host disease
  • allergic diseases including allergies, atopic diseases, allergic rhinitis;
  • bone diseases eg osteoporosis, Paget's disease
  • skin diseases including psoriasis, atopic dermatitis, UV(B)-induced dermal cell activation (eg sunburn, skin cancer);
  • gastrointestinal diseases including inflammatory bowel disease (including but not limited to ulcerative colitis, Crohn's disease), peptic ulceration, gastritis, oesophagitis, liver disease (including but not limited to cirrhosis, hepatitis).
  • MIF cytokine or biological activity is implicated in flic above diseases and conditions.
  • die disease or condition is selected from, the group consisting of rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, multiple sclerosis, psoriasis, uveitis, diabetes mellitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and chronic obstructive pulmonary disease.
  • the present invention provides a compound of Formula (II) or a 0 pharmaceutically acceptable salt or prodrug thereof wherein:
  • X is selected from - -O-, -S-, -C(R 5 )(R 5 -)- and -N(R 6 )-;
  • Y is selected from - -N(R 7 )-, -O-, and -S-;
  • R 1 is selected from hydrogen, Ci-C 3 alkyl, (CRsRsO 11 OR 7 , C(R 5 R 5 O n SR 7 , (CR 5 R 5 O I1 N(R G ) 2 and (CR s R 5 ')nhalo;
  • R 1 is selected from hydrogen, C r C ft alkyl, (CRi 0 R 10OpNR 14 RiS, (CR 1 eR 1 sOpOR ⁇ , (CR, 6 R 16 O p SR 17 , (CR 1 gR 1 eOphalo, (CR 16 R 1n OpNO 2 , (CR lft R Ift .) n C(O)R 2H , (CR lft R ]ft ) n S(O)R 17 , (CR 16 R 16 OaS(O) 2 R 17 , (CR 16 Ru) n S(O)SR 17 , and
  • R4 is selected from hydrogen, halogen, C
  • each R5 and R ? is independently selected from hydrogen, Ci-Cjalkyl, halo, OR 7 , SR 7 and N(Re) 2 ;
  • each R f is independently selected from hydrogen, Ci-C 3 alkyl and OR 7 ;
  • each R 7 is independently selected from hydrogen and Ci-C ⁇ alkyl
  • each R 12 and R 12' is independently selected from hydrogen, C 1 -C( J aIlCyI, C 2 -C ⁇ alke ⁇ yl, C 2 - C 6 alkynyl, OR 24 , SR 24 , halo, N(R 24 ⁇ , CO 2 R 24 , CN, NO 2 , aryl and heterocyclyl;
  • each R 1 4 and R15 is independently selected from hydrogen, CrC ⁇ alkyl, OR 17 , SR 1 7, and N(Rn) 2 ;
  • each R 16 and R 16 - is independently selected from hydrogen, C 1 -C 3 alkyl, halo, ORn, SRn and N(R 17 ) 2 ;
  • each R [ 7 is independently selected from hydrogen and Ci-C ⁇ alkyl
  • each R 1 S is independently selected from hydrogen and halo
  • R22 is selected from Ci-C 6 alkyl, NH 2 , NH(Ci-C 6 aikyl), N(Ci-C ⁇ alkyl) 2 , OR 29 or SR 20 ;
  • each R 24 is selected from H and Ci-C ⁇ alkyl
  • R28 is selected from hydrogen, Cj-Qalkyl, OR20, SR 2 0 or N(R 2 (J) 2 ;
  • Q is selected from O , S , S(0) u where u is an integer from 1 to 2;
  • R40 is selected from 11 7 OH, and C(R 4 IR 41 OvR 42 ;
  • each R 41 and R ⁇ ' is independently selected from H, OH, halo, NH 2 , CN and NO 2;
  • R 42 is selected from H, OR4.1, COOR 4 :!, CON(Ri 3 R 4S -), 0(CO)R 43 , aryl, and hcterocyelyl;
  • each R43 and R «- is independently selected from 11, Cj 0 alky], and benzyl;
  • n O or 1 to 3;
  • t is an integer from 1 to 10;
  • v is 0 or an integer from 1 to 10
  • the present invention provides a compound of Formula IH or a pharmaceutically acceptable salt or prodrug thereof wherein:
  • X is selected from - -O-, -S-, -C(R 5 )(Rs')- and -N(R 6 )-;
  • Y is selected from - -N(R 7 ), -O-, and -S-;
  • R 1 is selected from hydrogen, Crdalkyl, (CR 5 Ks ⁇ OR 7 , C(R 5 R 5 OnSR 7 , (CR 5 R 5 O n N(Re) 2 and (CR 5 R 5 ') ⁇ halo;
  • R 4 is selected from hydrogen, halogen, Cj-Qsalkyl, C 2 -C.ialkenyl, Cj-Cjalkynyl and (CR 12 Ri 2 On(CR 1 S) 3 ; .
  • each R 5 and Rj ' is independently selected from hydrogen, C]-Cjalkyl f halo, OR 7 , SR 7 and N(Re) 2 ;
  • each R f i is independently selected from hydrogen, C ⁇ -C 3 alkyl and OR 7 ;
  • each R ⁇ is independently selected from hydrogen and Q-Cjalkyl
  • each Rn and R12 ' is independently selected from hydrogen, Cj-C f talkyl, C2-C ⁇ alkenyl, C 2 - Cgalkynyl, OR 24 , SR 24 , halo, N(R ⁇ ) 2 , CO 2 R 24 , CN, NO 2 , ary! and heterocyclyl;
  • each R 14 and R 15 are independently .selected from hydrogen, C[-C 3 alkyl, OR 17 , SR 17 , and N(R 17 ) 2 ;
  • each R 16 and Rw is independently selected from hydrogen, Ci-Cjalkyl, halo, OR] 7 , SR 17 and N(Rn) 2 ;
  • each Rn is independently selected from hydrogen and Ci-C 3 alkyl
  • each R ⁇ « is independently selected from hydrogen and halo
  • R22 is selected from Cj-C ⁇ alkyl, NH 2 , NH(Ci -C ⁇ alkyl), N(C]-C ⁇ ;alJ ⁇ :yl)2, OR29 or SR20;
  • each R 24 is selected from I ( and Ci-C ⁇ alfcyl;
  • R 1 S is selected from hydrogen, Ci-Cealkyl, OR29, SK59 or N(R 29 ) ⁇ ;
  • each R 29 is independently selected from hydrogen and Q-Caalkyl
  • each R 45 and Rj 5 - is independently selected from H, OH, halo, NH 2 , CN, NO 2 ;
  • each R 46 is selected from COOR 47 , CON(R 47 R 47 '), 0(CO)R 47 , N(R 47 R 4 /);
  • each R 47 and R 47' is independently selected from H, C
  • R 4 ⁇ can be OR 47 ;
  • R ⁇ n can be H
  • n O or 1 10 3;
  • n O or an integer from 1 to 8;
  • p is O or an integer from 1 to 6;
  • t is an integer from 1 to 10;
  • v is O or an. integer from 1 to 10
  • a further aspect of the invention provides for the use of a compound of Formula (I) pr a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a. medicament for the treatment of a disease or condition as above.
  • a further aspect of the invention provides a pharmaceutical composition comprising a compound of the second or third aspect and a pharmaceutically acceptable carrier, diluent or excipient.
  • the present invention provides a method of inhibiting cytokine or biological activity of MIF comprising contacting MIF with a cytokine or biological inhibiting amount of a compound of formula (I), or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention provides a method of treating, preventing or diagnosing a disease or condition wherein MfF cytokine or biological activity is implicated comprising the administration of a treatment, prevention or diagnostic effective amount of a compound of formula (T) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof.
  • the invention provides a method of treating or preventing a disease or condition wherein MIF cytokine or biological activity is implicated comprising:
  • the present invention provides a method of prophylaxis or treatment of a disease or condition for which treatment with a glucocorticoid is indicated, said method comprising:
  • the present invention provides a method of treating steroid-resistant administering to a mammal a glucocorticoid and a compound of formula (I).
  • the present invention provides a method of enhancing the effect of a glucocorticoid in mammals comprising administering a compound of formula (I) simultaneously, separately or sequentially with said glucocorticoid.
  • the present invention provides a pharmaceutical composition comprising a glucocorticoid and a compound of formula (I).
  • a glucocorticoid in the manufacture of a medicament, for administration with a compound of formula (I) for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
  • a compound of formula (I) in the manufacture of a medicament for administration with a glucocorticoid for the treatment or prophylaxis of a disease or condition for which treatment of a glucocorticoid is indicated.
  • a glucocorticoid and a compound of formula (T) in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
  • Inhibitors of MIF may also be used in implantable devices such as stems. Accordingly, in a further aspect the present invention provides an implantable device, preferably a stent, comprising;
  • the present invention provides a method of treating, preventing or diagnosing a disease or condition wherein MIF cytokine activity is implicated comprising the step of implanting an implantable device according to the invention in a subject in need thereof.
  • the present invention further provides an angioplaslic stent for inhibiting the onsel of restenosis, which comprises an angioplasty stent operably coated with a prophylactically effective dose of a composition comprising at least o ⁇ e.compound of formula (I).
  • the present invention further provides a method for inhibiting the onset of restenosis in a subject undergoing angioplasty, which comprises topically administering a stent according to the present invention to the subject at around the time of the angioplasty.
  • Figure I shows that treatment with a compound according to the present invention induces a dose-dcpende ⁇ t inhibition of LPS-induced IL-6 production in a mouse macrophage cell line.
  • Figure 2 shows that treatment with a compound according to the present invention induces a dose-dependent inhibition of IL-I induced COX-2 expression when S112 cells are treated with up to 100 ⁇ lvl concentration of compound.
  • Figure 3A shows that treatment of mice with compound 15 according to the present invention results in a significant dose-dependent suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock.
  • Figure 3B shows that treatment of mice with compounds 2 and 13 according to die present invention results in a significant dosc-dependenl suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock.
  • Figure 30 shows that treatment of mice with compound 4 according io the present invention results in a significant dose-dependent suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock.
  • Figure 3D shows that treatment of mice with compound 19 according to the present invention results hi a significant dose-depe ⁇ dcnt suppression of LPS-induccd scrum TNF levels in a mouse model of endotoxic shock.
  • Figure 4 shows reduction in DTH reactions in vivo in mice treated with compound 13.
  • Figure 5 shows effect of compound 13 on rhMIF-induced leukocyte adhesion
  • the present invention provides a method of treating, diagnosing or preventing autoimmune diseases, tumours, or chronic or acute inflammatory diseases comprising administering a treatment, prevention or diagnostic effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof wherein:
  • X is selected from - -O-, -S-, -C(R 5 )(R 5 )- and -N(R 0 )-;
  • Y is selected from - -N(R 7 )-, -O 7 -S-, and -C(Ry) 2 -;
  • R 1 is selected from hydrogen, Ci-C 3 alkyl, (CR 5 R 5 O n OR 7 , C(RsRsO n SR 7 , (CR 5 R 5 OnN(Ro) 2 and (CRsRsOnhalo;
  • R 3 is selected from hydrogen, Ci-Qalkyl, (CR 16 R 1 n O n NR 11 IR 15 , (CRI 6 RJ 0 O P ORI 7 , (CR 1 ftR 1 ⁇ OpSR ⁇ , (CR, ti R ⁇ 6 )phalo, (CR 1 oR 1 dOpNOj, (CR 16 R 10 O n C(O)R 2 R, (CR 1n R 16 O 11 S(O)R 17 , (CR 1n R 16 OnS(O) 2 R 17 , (CRK 1 R 16 O 1I S(O) 3 R 17 , and
  • R 4 is selected from hydrogen, halogen, Ci-Cjalkyl, C 2 -Csalkenyl, C 2 -C3alkynyl and
  • each R 5 and Ry is independently selected from hydrogen, Ci-Cjalkyl, halo v OR 7 , SR7 and N(Re) 2 ;
  • each R 6 is independently selected from hydrogen, Ci-Cjalkyl and ORy;
  • each R 7 is independently selected from hydrogen and Ci-Cgalkyl
  • each R 12 and R 12 * is independently selected from hydrogen, C
  • each R 14 - UId R i 3 is independently selected from hydrogen, Ci-C 3 alkyl, ORn, SRi 7 , and N(R 17 ⁇ ;
  • each R 1 ⁇ and R 1n ' is independently selected from hydrogen, C]-C 3 alkyl, halo, OR 17 , SR 17 and N(Rn) 2 ;
  • each Rn is independently selected from hydrogen and Ci-Cjalkyl
  • each R 1 g is independently selected from hydrogen and halo
  • R 22 is selected from d-Qalkyl, NH 2 , NH(C,-C 6 alkyl), N(C,-C6al.kyl)2, OR 2 y or SR 29 ; each R 24 is selected from H and Ci-Cgalkyl;
  • R 2 ⁇ is selected from hydrogen, Ci-C(jaLkyl, OR 2 9, SR29 or N(R 29 ) 2 ;
  • each R 2 O is independently selected from hydrogen and Ci-Csalkyl
  • Q is selected from O, S, NR 40 , S(O) 11 where u is an integer frornl to 2;
  • R 40 is selected from H, OH, and
  • each R 41 and R 4T is independently selected from H, OH, halo, NH 2 , eyan ⁇ , and NO 2 ;
  • R 12 is independently selected from H, OR43, COOR 4 J, CON(R 43 R 4 .?), 0(CO)R 43 , aryl, and heterocyclyl;
  • each R43 find R 43' is independently selected from H, Ci . ⁇ j ⁇ ilkyl, benzyl, and aryl;
  • n O or an integer to 3
  • n O or an integer from I to 20;
  • p is 0 or an integer from 1 to 6;
  • t is an integer from 1 to 10
  • v is 0 or an integer from 1 to 10.
  • the autoimmune disease, tumour, or chronic or acute inflammatory disease is selected from the group comprising:
  • rheumatic diseases including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis
  • spondyloarthropathies including but not limited to ankylosing spondylitis, reactive arthritis, Reiter's syndrome
  • crystal arthropathies including but not limited to gout, pseudogout, calcium pyrophosphate deposition disease
  • Lyme disease polymyalgia rheumatica
  • connective tissue diseases including but not limited to systemic lupus erythematosus, systemic sclerosis, polymyositis, derruatomyositis, Sjogren's syndrome
  • connective tissue diseases including but not limited to systemic lupus erythematosus, systemic sclerosis, polymyositis, derruatomyositis, Sjogren's syndrome
  • vasculitides including but not limited to polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome;
  • vascular diseases including atherosclerotic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischaemic heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis;
  • ocular diseases including uveitis, corneal disease, ulcerative colitis, cataracts; autoimmune diseases (including but not limited to diabetes mellitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis);
  • pulmonary diseases including but not limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome;
  • cancers whether primary or metastatic including but not limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
  • renal diseases including glomerulonephritis, interstitial ncpliritis;
  • hypothalamic-pituitary-adrenal axis disorders of the hypothalamic-pituitary-adrenal axis
  • nervous system disorders including multiple sclerosis, Alzheimer's disease; rheumatoid arthritis, cancer), endometrial function (menstruation, implantation, endometriosis);
  • infective disorders including cndotoxic (septic) shock, cxotoxic (septic) shock, infective (true septic) shock, malarial complications, other complications of infection, pelvic inflammatory disease;
  • transplant rejection graft-versus-host disease
  • allergic diseases including allergies, atopic diseases, allergic rhinitis;
  • bone diseases eg osteoporosis, Paget's disease
  • skin diseases including psoriasis, atopic dermatitis, UV(B)-induced dermal cell acLivalion (eg sunburn, skin cancer);
  • gastrointestinal diseases including inflammatory bowel disease (including but not limited to ulcerative colitis, Crohn's disease) * peptic ulceration, gastritis, oesophagitis, liver disease (including but not limited to cirrhosis, hepatitis).
  • MIF cytokine or biological activity is implicated in the above diseases and conditions.
  • flic disease or condition is selected from the group consisting of rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, multiple sclerosis, psoriasis, uveitis, diabetes mellitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and chronic obstructive pulmonary disease.
  • R 40 is C(RnR-H OvRa wherein R.,2 is COOR43. More preferably, R 43 is hydrogen or Ci-C ⁇ alkyl, preferably methyl.
  • lhe compound of Formula 1 is selected from any one of Compounds 1 to 32 as set out in Lhe Examples herein.
  • the term "effective amount" relates to an amount of compound which, when administered according to a desired dosing regimen, provides the desired MIF cytokine inhibiting or treatment or therapeutic activity, or disease/condition prevention. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods.
  • a cytokine or biological activity inhibiting amount is an amount which will at least partially inhibit the cytokine or biological activity of MIF.
  • a therapeutic, or treatment, effective amount is an amount, of the compound which, when administered according to a desired dosing regimen, is sufficient to at least partially attain the desired therapeutic effect, or delay the onset of, or inhibit the progression of or halt or partially or fully reverse the onset or progression of a particular disease condition being treated.
  • a prevention effective amount is an amount of compound which when administered according to the desired dosing regimen is sufficient to at least partially prevent or delay the onset of a particular disease or condition.
  • a diagnostic effective amount of compound is an amount sufficient to bind to !VtIF to enable detection of the MIF-compound complex such that diagnosis of a disease or condition is possible.
  • Suitable dosages may lie within the range of about 0.1 ng per kg of body weight to 1 g per kg of body weight per dosage.
  • the dosage is preferably in the range of 1 ⁇ g to 1 g per kg of body weight .per dosage, such as is in the range of 1 mg to 1 g per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 500 mg per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 250 mg per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per kg of body weight per dosage.
  • the dosage is in the range of l ⁇ g to lmg per kg of body weight per dosage.
  • Suitable dosage amounts and dosing regimens can be determined by the attending physician or veterinarian and may depend on the desired level of inhibiting activity, the particular condition being treated, the severity of the condition as well as the general age, health and weighL of lhe subject.
  • the active ingredient may be administered in a single dose or a series of doses. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition.
  • therapeutically active agents such as anti-inflammatory (eg steroids such as glucocorticoids) or anti-cancer agents may be used in conjunction with a compound of Formula (I).
  • Compounds of Formula (1) when administered in conjunction with other therapeutically active agents may exhibit an additive or synergistic effect. These may be administered simultaneously, either as a combined form (ie as a single composition containing the active agents) or as discrete dosages. Alternatively, the other therapeutically active agents may be administered sequentially or separately with the compounds of the invention.
  • the invention also relates to kits and combinations, comprising a compound of Formula (I) and one or more other therapeutically active ingredients for use in the treatment of diseases or conditions described herein.
  • agents which could be used in combination with a compound of Formula (I) include: antirheumatic drugs (including but not limited to methotrexate, leflunomide, sulphasalazi ⁇ e, hydroxycholorquine, gold salts); immunosuppressive drugs (including but not limited to cyclosporin, mycophenyllate mofet ⁇ , azathioprine, cyclophosphamide); anti-cytokine therapies (including but not limited to antagonists of, antibodies to, binding proteins for, or soluble receptors for tumor necrosis factor, interleukin 1, interleukin 3, intcrlcukin 5, interleukin 6 ⁇ interleukin 8, interleukin 12, interleukin 18, interleukin 17, and other pro-inflammatory cytokines as may be found relevant to pathological states); antagonists or inhibitors of mitogen-activated protein (MAP) kinases (including but not limited to antagonists or inhibitors of extracellular signal-regulated kinases (ERMAP) kinases
  • the compound of formula (I) is administered in conjunction with a second therapeutic agent.
  • the second therapeutic agent is a glucocorticoid.
  • the compound of Formula (I) is a compound of Formula (TI) wherein:
  • ⁇ X is selected from - -O-, -S-, -C(R 5 )(RsO- and -N(R 6 )-;
  • R 1 is selected from hydrogen, Ci-G*alkyl, (CR 5 R 5 ⁇ OR 7 , C(RsRsOnSR?, CCR 5 Ry) n N(Rs) 2 and (CRsRsOnhalo;
  • R 3 is selected from hydrogen, C 1 -C 6 QIlCyI, (CR] 6 R 16 OpNR 14 R 1 S, (CR 1ft R 1 ⁇ -) P OR, 7 , (CR 16 R 1 ft .)pSR 1 7, (CR 1 6R 1 ff)phalo, (CR 1n R 1n O n NO 2 , (CR 16 R 16 ⁇ C(O)R 28 , (CR 16 R 16 OnS(O) 2 Ri 7 , (CRi 6 RKy) n S(O) 3 R 17 , and (CR ⁇ R ⁇ ff )pC(R 1 8)j;
  • R 4 is selected from hydrogen, halogen, C ⁇ -C 3 alkyl, C2-C 3 alk.en.yl, C 2 .-C 3 alky-.yl and (CR 12 RJr) n (CR 1 B) 3 ;
  • each R ⁇ and Rs> is independently selected from hydrogen, Ci-C/jalkyl, halo, OR 7 , SR 7 and N(R ⁇ ) 2 ;
  • each Rs is independently selected from hydrogen, Ci-Csalkyl and OR 7 ;
  • each R 7 is independently selected from hydrogen and Ci-Cjalkyt
  • each R 12 and R ⁇ - is independently selected from hydrogen, Ci-C ⁇ alkyl, C 2 -Coalkenyl, C 2 - C ⁇ alkynyl, OR 2 /), SR24, halo, N(R ⁇ ) 2 , CO 2 R 24 , CN, NO 2 , aryl and heterocyclyl;
  • each R 14 and R 15 is independently selected from hydrogen, CrC 3 alkyl, OR) 7 , SRn, and
  • each R 1 g and RK,- is independently selected from hydrogen, C ⁇ -C 3 _ ⁇ lkyl, halo, OR 17 , SR n and N(R 1 V) 2 ;
  • each R 17 is independently selected from hydrogen and C ⁇ -C 3 alkyl
  • each R 1 g is independently selected from hydrogen and halo
  • R 22 is selected from Q-Qalkyl, NH 2 , NH(Cj-C 6 al]cyl), N(Ci-C6alkyl) 2 , OR 2 y or SR 2i ,; each R 24 is selected from H and Ci-Cr,alkyl;
  • R 2K is selected from hydrogen, Cj-Cealkyl, OR25,, SR 2 9 «r N(R ⁇ h;
  • each R 2 9 is independently se lected from hydrogen and C 1 -C ⁇ alkyl
  • Q is selected from O , S , S(O) 11 where u is an integer from 1 to 2;
  • R 4 0 is selected from H, OH, and C(R ⁇ R4j')vR42;
  • each R 41 and E 4 i- is independently selected from H. OH, halo, NH2, CN and NO2;
  • R ⁇ 2 is selected from H, OR 4 :), COOR43, CON(R 43 R 43 .), O(CO)R 43 ⁇ N(R 4 ⁇ R43'), aryl, and heterocyelyl;
  • each R/i; ⁇ and R 43 - is independently selected from H, Ci-ealkyl, and benzyl;
  • n O or l to 3;
  • n O or an integer from 1 to 8;
  • p is O or an. integer from 1 to 6;
  • t is an integer from 1 to ,10;
  • v is 0 or an integer from 1 to 10.
  • the compound of Formula (I) is a compound of Formula (III) wherein:
  • X is selected from - -O-, -S-, -C(R 5 )(R 5 ')- and -N(R fi )-;
  • Y is selected from - -N(R 7 )-, -O-, and -S-;
  • R 1 is selected from hydrogen, C,-C 3 alkyl, (CR 5 RSO E OR 7 , C(R 5 R 5 ⁇ SR 7 , (CRSR 5 OJN(R( V )J and (CR 5 R 5 O n IIaIo;
  • R 4 is selected from hydrogen, halogen, d-Cjalkyl, Cz-C ⁇ alkenyl, C 2 -Cjalkyriyl and (CRuR 12 On(CR 1 S) 3 ;
  • each Rj and Ry is independently selected from hydrogen, Ci-C 3 alkyl, halo, OR 7 , SR 7 and N(R ⁇ ) 2 ;
  • each R f1 is independently selected from hydrogen, Ci-C ⁇ alkyl and OR 7 ;
  • each R7 is independently selected from hydrogen and CrQ ⁇ aLkyl
  • each R 12 and R 12 ' is independently selected from hydrogen, C ⁇ -C 6 -alkyl, C 2 -C ⁇ -dkcnyl, C 2 - Cealkynyl, OR 24 , SR 24 , halo, N(R 24 ) ⁇ , CO 2 R 2 /!, CN, NO 2 , aryl and heterocyclyl;
  • each Ru and R 15 are independently selected from hydrogen, Ci-C ⁇ alkyl, OR 17 , SR 17 , and N(Rn) 2 ;
  • each R] 0 and R 1 ⁇ is independently selected from hydrogen, Ci-C-jalkyl, halo, OR 17 , SR 17 and N(R 17 J 2 ; each Rn is independently selected from hydrogen and C)-C 3 alkyl;
  • each R 1 s is independently selected from hydrogen and halo
  • R 22 is selected from C,-C 6 alkyl, NH 2 , NH(Q-C 6 alkyl) ⁇ N(C r C 6 alkyl) 2 , OR 29 Or SR 29 ;
  • each R ⁇ 4 is selected from H and C
  • R2 8 is selected from hydrogen, CrC ⁇ alkyl, OR 29 , SRM or N(R ⁇ ) 2 ;
  • each R 2 9 is independently selected from hydrogen and Ci-C 3 alkyl
  • R44 is selected from OH
  • each R45 and R 4 S' is independently selected from H, OH, halo, NH 2 , CN, NO 2 ;
  • each R/jft is selected from COOR 4 ?, CON(R 47 R 47 ), 0(CO)R 47 , N(R 47 R 47 );
  • each R 17 and R47 ' is independently selected from H, Cus alkyl, benzyl;
  • R 4 g can be 11;
  • is U or 1 to 3;
  • n O or an integer from 1 to 8;
  • p is O or an integer from 1 to 6;
  • t is an integer from 1 to 10;
  • v is O or an integer from 1 to 10.
  • the present invention provides a compound of Formula (II) or a pharmaceutically acceptable suit or prodrug thereof wherein:
  • X is selected from - -O-, -S-, -C(R 5 )(R 5 1 )- md -N(R n )S
  • Y is selected from - -N(R 7 )-, -O-, and -S-;
  • R 1 is selected from hydrogen, C,-C 3 alkyl, (CR 5 R 5 O H OR 7 , C(R 5 R 5 ⁇ SR 7 , (CR 5 RsOnN(Rn) 2 and (CR 5 R 5 -X 1 IIaIo;
  • R; ⁇ is selected from hydrogen, Ci-C 6 alky], (CR 1n R 16 OpNR 14 R 1 S, (CR K ,R 16 0pQR-i7, (CR w R 1 «0pSR 17 , (CR 16 R 1ff ) p halo, (CR 16 R 16 OpNO 2 , (CR 16 Rw) 1 AOJR 28 , (CRi6R 1 6-)aS(O)R 1 7, (CR 1n R 16 OnS(O) 2 R 17 , (CR 10 R 1n O n S(O) 3 R 17 , and (CR 16 R 1 COpC(R 1 S).);
  • R 4 is selected from hydrogen, halogen, C
  • each Rs and Ry is independently selected from hydrogen, Ci-C3alkyl, halo, OR 7 , SR 7 and N(Re) 2 ;
  • each R 6 is independently selected from hydrogen, C
  • each R ⁇ is independently selected from hydrogen and Q-Csalkyl; each R 12 and R ⁇ is independently selected from hydrogen, Ci-C ⁇ lkyl, C 2 -C 6 alkenyl, Ca- C ⁇ alkyny ] , OR 24 , SR 24 , halo, N(R 24 )., CO 2 R 24 . CN, NO 2 , aryl and heterocydyl; .
  • each Ru and R15 is independently selected from hydrogen, Cj-Osalkyl, OR 17 , SRw > and N(R, 7)2;
  • each is independently selected from hydrogen, Ci-Csalkyl, halo, OR) 7 , SR 17 and
  • each R. J 7 is independently selected from hydrogen and Ci-Cjalkyl
  • each R 1 g is independently selected from hydrogen and halo
  • R 22 is selected from C r C ⁇ jalkyt, NH 21 NH(C ⁇ -C 6 alkyI), N(Ci-C ⁇ 5 ali.yl) 2 , OR 2 y or SR 29 ;
  • each R 24 is selected from H and Ci -Cgalkyl
  • Ra ? is selected from hydrogen, Cj-Coalkyl, OR 2 c», SR29 or N(R 29 ) 2 * ,
  • each R 2 ⁇ is independently selected from hydrogen and Ci-Caalkyl
  • Q is selected from O 1 S , S(O) U where u is an integer from 1 to 2;
  • R40 in selected from H, OH, and C(R4iR-n-)vR42;
  • each R 41 and R 41' is independently selected from H, OH, halo, NHo, CN and NO 2 ;
  • R 42 is selected firom H, OR 43 , COOR 43 , CON(R 4 ⁇ R 43 0, 0(CO)R 4 S, N(R 4 ⁇ y), aryl, and heterocyclyl; .
  • each R 43 and R*r is independently selected from H, Cut, alkyl, and benzyl;
  • n O or 1 to 3;
  • 0 m is O or an integer from ] to 8; p is ⁇ or an integer from 1 to 6;
  • t is an integer from 1 to 10;
  • v is 0 or nn integer from 1 to 10
  • the present invention provides a compound of Formula III or a pharmaceutically acceptable salt or prodrug thereof wherein:
  • X is selected from - -O-, -S-, -C(Rs)(R 5' )- and -N(R 6 )-;
  • Y is selected from - -N(R 7 )-, -O-, and -S-;
  • R 1 is selected from hydrogen.
  • R 3 is selected from hydrogen, Cj-Qalkyl, (CR 1 ⁇ R 1 fiOpNR ⁇ R 1 s, (CR 16 R I 6 OpOR n .
  • (CR ⁇ R 1 ⁇ O ⁇ SRn, (CR ⁇ R 1 sOphalo, (CR 15 R 16 O p NO 2 , (CR 16 R ⁇ ft -)nC(O)R 2 s, (CR 16 RIn) 1 A NR 24 )R 22 , (CR 16 R 16 OS(O)R 17 , (CR 1 ⁇ w) n S(O) 2 R 17 .
  • R 4 is selected from hydrogen, halogen, C,-C 3 alkyl, Ci-C ⁇ alkenyl, Cz-Cjatky ⁇ yl and (CR 12 R n O 11 (CR 18 )Ir,
  • each R,i and R5 1 is independently selected from hydrogen, C 1 -C 3 HUCyI, halo, OR 7 , SR 7 and N(R 6 ) 2 ;
  • each Ra is independently selected from hydrogen, Ci-Cjalkyl and OR 7 ;
  • each R 7 is independently selected from hydrogen and Ci-Cjalkyl
  • each R 1 2 and R 12' is independently selected from hydrogen, Cj-CoaLkyl, C ⁇ -Cgalkcnyl, C 2 - Cftalkynyl, OR 2 ⁇ , SR2 4 , halo, N(R3 4 ) 2 , CO 2 R 14 , CN, NO 2 , aryl aud heterocyclyl;
  • each RK and R 1S are independently selected from hydrogen, Ci-C 3 a!kyt, OR 17 , SR 17 , and N(R 17 )U
  • each R 1 g and R 1n - is independently selected from hydrogen, Ci-Qjalkyl, halo, OR 17 , SR 17 and N(R I7 ) 2 ⁇
  • each R 17 is independently selected from hydrogen and Ci-C- 3 alkyl
  • each R 1 x is independently selected from hydrogen and halo
  • R22 is selected from C
  • each R2/1 is selected from H and d-Qalkyl
  • R 2 H is selected from hydrogen, Ci-C o alkyl, OR 29 , SR 2 y or N(R 29 h;
  • each R. 2 9 is independently selected from, hydrogen and Ci-C 3 alkyl
  • R 44 is selected from OH
  • each R 4n is selected from COOR47, CON(R 47 R 47 '), 0(CO)R 47 , N(R 47 R 4? ');
  • each R 47 and R 47' is independently selected from H, Ci -6 af kyl, benzyl;
  • R 46 can be OR 47;
  • R 4 ⁇ can be H
  • n O or 1 to 3;
  • n O or an integer from 1 to 8;
  • p is O or an integer from Ho 6;
  • t is an integer from. 1 to 10;
  • v is 0 or an integer from 1 to 10
  • alkyl refers to monovalent straight, branched or, where appropriate, cyclic aliphatic radicals, having 1 to 3, I to 6, 1 to 10 or 1 to 20 carbon atoms, e.g. methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, ⁇ -butyl, sec-butyl, t-butyl and cyclobutyl, n-pentyl, 1-methylbutyl, 2-methylb ⁇ tyl, 3-methylbutyl, cyclopcntyl, n-hcxyl, 1- 2- 3- or 4- methylpcntyl, 1- 2- or 3-cthylbulyl, 1 or 2- propylpropyl or cyclohexyl.
  • An alkyl group may be optionally substituted one or more times by halo (eg chloro, lluoro or bromo), CN, NO 2 , CO 2 H, CQA- ⁇ alkyl, CO 2 NH 2 , CO 2 NH(C !-fi alkyl), CO 2 N(C I-6 aIkyl) 2 , OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl , benzyloxy, phenoxy, NH 2 ,
  • halo eg chloro, lluoro or bromo
  • alkoxy examples include methoxy, ethoxy, n-propoxy, iso-propoxy, cyclopropoxy, and butoxy (n-, sec- 1- and cyclo) pentoxy and hexyloxy.
  • alkoxy examples include methoxy, ethoxy, n-propoxy, iso-propoxy, cyclopropoxy, and butoxy (n-, sec- 1- and cyclo) pentoxy and hexyloxy.
  • the "alkyl" portion of an alkoxy group may be substituted as described above,
  • alkenyl refers to straight, branched, or where appropriate, cyclic carbon containing radicals having one or more double bonds between carbon atoms.
  • radicals include vinyl, allyl, butcnyl, or longer carbon chains such as those derived from palmitoleie, oleic, linoleic, linolenic or arachidonic acids.
  • An alkenyl group may be optionally substituted one or more tunes by hal ⁇ (eg chloro, f luoro or bromo), CN, NO 2 , CO 2 H, CO 2 NH 2 , CO 3 NH(C 1 6 alky0, CO 2 N(C J-s alkyl) 2 , OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl, benzyloxy, phenoxy, NHi, or N(Ci-SaUCyI) 2 .
  • ⁇ prelarred optional substituent is a polar substituent.
  • alkynyl refers to straight or branched carbon containing radicals having one or more triple bonds between carbon atoms. Examples of such radicals include propargyl, butynyl and hexynyl.
  • An alkynyl group may be optionally substituted one or more times by halo (eg chloro, fluoro or bromo), CN, NO 2 , CO 2 H, COaNH 2 , CQ 2 NH(C 1 .6alkyl), OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl, benzyloxy, phenoxy.
  • halo eg chloro, fluoro or bromo
  • Suitable NI I(alkyl) and N(alkyl> 2 include methylamino, ethylamino, isopropylami ⁇ o, dimethylami ⁇ o, n-propylamino, diethylamino and di-isopropylamino.
  • halogen refers to fluorine (fluoro), chlorine (chloro), bromine (bromo)
  • An aryl gtoup refers to C ⁇ -Cio aryl groups such as phenyl or naphthalene.
  • Aryl groups may be optionally substituted one or more times by halo (eg, chloro, fluoro or bto ⁇ ), CN, NO 2 , CO 2 H, CO 2 Ci -6 alkyl, CO 2 NH 2 , COnNH(C 1 .
  • heterocyclyl refers to a cyclic, aliphatic or aromatic radical containing at least one heteroatom independently selected from O, N or S
  • suitable heterocyclyl groups include fury], dioxolanyl, dioxanyl, dithiaiiyl, dithiolanyl, pyridinyl, pyrimidinyl, pyrazolyl, piperidiiiyl, pyrrolyl, thyaphenyl, oxazotyl, imidazolyl, thjazolyl, ⁇ soxazolyl, isothiazolyl, quinolyl, isoquinolyl, indolyl, betLt ⁇ furanyl, benzolhiophenyl, triazolyl, tetrazolyl, oxadiazolyl and puri ⁇ yl.
  • Heterocyclyl groups may be optionally substituted one or more times by halo (eg, cliloro, fluoro or bromo), CN, NO 2 , CO 2 H, CO 2 C ⁇ alkyl, CO 2 NII 2 , CO 2 NH(Ci. 6 alkyl), CO 2 N(C,. ⁇ alkyl) 2j OH, alkoxy, acyl, acetyl, halomethyl, trifluoromelhyl, benzyloxy, phenoxy, NH 2 , NH(Ci -salkyl) or N(C 1-fi aIkyl) 2 .
  • halo eg, cliloro, fluoro or bromo
  • salt, or prodrug includes any pharmaceutically acceptable salt, ester, solvate, hydrate or any oilier compound which, upon administration to the recipient is capable of providing (directly or indirectly) a compound of Formula (1) as described herein.
  • pro-drug is used in its broadest sense and encompasses those derivatives that, arc converted in vivo to the compounds of the invention. Such derivatives would ⁇ eadily occur to those skilled in the art, and include, for example, compounds where a free hydroxy group is converted into an ester, such as an acetate, or where a free amino group is converted into an amide.
  • Procedures for acylating hydroxy or amino groups of the compounds of the invention are well known in the art and may include treatment of the compound with an appropriate carboxylic acid, anhydride or acylchloridc in the presence of a suitable catalyst or base.
  • Suitable pharmaceutical Iy acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, maleic, citric, lactic, muck, gluconic, benzoic, succinic, oxalic, phenyl acetic, methanesulphonic, tolucnesulphonic, benezenesulphonie, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
  • pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric,
  • Base salts include, but are not limiled to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylanimonium.
  • Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • lower alkyl halide such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl and diethyl sulfate; and others.
  • the invention thus also relates to compounds in substantially pure isomeric form at one or more asymmetric centres eg., greater than about 90% cc, such as about 95% or 97% ee or greater than 99% ce, as well as mixtures, including racemic mixtures, thereof.
  • Such isomers may be prepared by asymmetric synthesis, for example using chiral intermediates, or by chiral resolution.
  • ⁇ further aspect of the invention provides for the use of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of a disease or condition as above.
  • a pharmaceutical composition comprising a compound of formula. (I) together with a pharmaceutically acceptable carrier, diluent or excipie ⁇ t.
  • compositions of such compositions may contain pharmaceutically acceptable additives such as carriers, diluents or excipicnts. These include, where appropriate, all conventional solvents, dispersion agents, fillers, solid earners, coating agents, antifungal and antibacterial agents, dermal penelralion agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
  • pharmaceutically acceptable additives such as carriers, diluents or excipicnts. These include, where appropriate, all conventional solvents, dispersion agents, fillers, solid earners, coating agents, antifungal and antibacterial agents, dermal penelralion agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
  • compositions include those suitable for oral, rectal, Lnhalational, nasal, transdermal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intraspinal, intravenous and intradermal) administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions for use in the present invention may be formulated to be water or lipid soluble.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in- water liquid emulsion or a watcr-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder ov granules, optionally mixed with a binder (eg inert diluent, preservative, disinlegra ⁇ t (eg. sodium starch glyc-olate, cross-linked polyvinyl ovrrolidone, cross-linked sodium carboxvr ⁇ elhvl cellulose 1 ) 1 ) surface- active or dispersing agent.
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • compositions suitable for topical administration in the mouth iaclude lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • the compounds of Formula (1) may also be administered intranasal Iy or via inhalation, for example by atomiser, aerosol O ⁇ nebulizer means.
  • compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like.
  • suitable carriers include mineral oil, propylene glycol, polyoxycthylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodeeanol, benzyl alcohol and water.
  • Transdermal devices such as patches, may also be used to administer the compounds of the invention.
  • compositions for rectal administration may be presented as a suppository with a suitable carrier base comprising, for example, cocoa butter, gelatin, glycerin or polyethylene glycol.
  • compositions suitable for vaginal admin istration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which, render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • sterile liquid carrier for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from slerilc powders, granules and tablets of lhe kind previously described.
  • Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the active ingredient.
  • compositions of this invention may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents, disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents.
  • suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine.
  • Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xa ⁇ than gum, bentonitc, alginic acid or agar.
  • Suitable flavouring agents include peppermint oil, oil of wivettergreen, cherry, orange or raspberry flavouring.
  • Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zcifl, shellac or gluten.
  • Suitable preservatives include sodium bcnzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite.
  • Suitable lubricants include magnesium stearate, stearic acid, sodium oleatc, sodium chloride or talc.
  • Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
  • the present invention provides a method of inhibiting cytokine or biological activity of MIF comprising contacting MIK with a cytokine or biological activity inhibiting effective amount of a compound of formula (I), or a pharmaceutically acceptable salt Of prodrug thereof.
  • the invention provides a method of treating, preventing or diagnosing a disease or condition wherein MlF cytokine or biological activity is implicated comprising the administration of a treatment, prevention or diagnostic effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof.
  • a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment, prevention or diagnosis of a disease or condition wherein MlF cytokine or biological activity is implicated.
  • MIF includes liuina ⁇ or other animal MlF and derivatives and naturally occurring variants thereof which at least partially retain MIF cytokine or biological activity.
  • the subject to be treated may be human or other animal such as a mammal.
  • Non-human, subjects include, but are not limited to primates, livestock animals (eg sheep, cows, horses, pigs, goats), domestic animals (eg dogs, cats), birds and laboratory test animals (eg mice. rats, guinea pigs, rabbits).
  • JMHF is also expressed in plants (thus "MIF" may also refer to plant MIF) and where appropriate, compounds of Formula (I) may be used in botanical/agricultural applications such as crop control.
  • cytokine or biological activity of MIF includes Lhe cytokine or biological effect on cellular function via autocrine, endocrine, paracrine, cytokine, hormone or growth factor activity or via intracellular effects.
  • -he invention provides a method of treating or preventing a disease or condition wherein MIF cytokine or biological activity is implicated comprising:
  • the second therapeutic agent Is a glucocorticoid compound.
  • the present invention provides a method of prophylaxis or treatment of a disease or condition for which treatment with a glucocorticoid is indicated, said method comprising: administering to a mammal a glucocorticoid and a compound of formula (1).
  • the present invention provides a method of treating steroid-resistant diseases comprising administering to a mammal a glucocorticoid and a compound of formula (I).
  • the present invention provides a method of enhancing the effect of a glucocorticoid in mammals comprising administering a compound of formula (I) simultaneously, separately ov sequentially with said glucocorticoid.
  • the present invention provides a composition comprising a glucocorticoid and a compound of formula (I).
  • a glucocorticoid in the manufacture of a medicament for administration with a compound of formula (I) for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
  • a compound of formula (I) in the manufacture of a medicament for administration with a glucocorticoid for the treatment or prophylaxis of a disease or condition for which, treatment of a glucocorticoid in indicated.
  • glucocorticoid and a compound of formula (I) in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
  • the amount of glucocorticoid used in the methods, uses and compositions of the invention is less than the amount which would he effective ⁇ a the absence of the compound of formula (I).
  • any amount of glucocorticoid which is effective in combination with a compound of formula (I) is considered less than the amount which would be effective in the absence of a compound formula (I). Accordingly, the invention provides a steroid-sparing therapy.
  • disease or condition for which treatment with a glucocorticoid is indicated refers to diseases or conditions which are capable of being treated by admmistraiion of a glucocorticoid including but not limited to autoimmune diseases, tumours, or chronic or acute inflammatory diseases. Examples of such diseases or conditions include:
  • rheumatic diseases including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis
  • spondyloarthropathies including but not limited to ankylosing spondylitis, reacLive arthritis, Reiter's syndrome
  • crystal arthropathies including but not limited to gout, pscudogoul, calcium pyrophosphate deposition disease
  • Lyme disease polymyalgia rheumatica
  • connective tissue diseases including but not limited to systemic lupus erythematosus, systemic sclerosis, polymyositis, dermatomyositis, Sjogren's syndrome;
  • vasculilides including but not liir ⁇ Ledto polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome);
  • vascular diseases including atherosclerotic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischacniie heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis; 43.
  • autoimmune diseases including but not limited to diabetes mellitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis;
  • pulmonary diseases including but not limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome;
  • cancers whether primary or metastatic including but not limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
  • renal diseases including glomerulonephritis, interstitial nephritis;
  • hypothalamic-pituitary-adrenal axis disorders of the hypothalamic-pituitary-adrenal axis
  • nervous system disorders including multiple sclerosis, Alzheimer's disease;
  • angiogcncsis eg diabetic retinopathy, rheumatoid arthritis, cancer
  • endometrial function e.g., implantation, endometriosis
  • transplant rejection graft- vcrsus-host disease
  • allergic diseases including allergies, atopic diseases, aHeTgic rhinitis;
  • bone diseases eg osteoporosis, Pagct's disease
  • skin diseases including psoriasis, atopic dermatitis, UV(B)-induccd dermal cell activation (eg sunburn, skin cancer); pain, testicular dysfunctions and wound healing;
  • gastrointestinal diseases including inllammaiory bowel disease (including but not limited to ulcerative colitis, Crohn's disease), peptic ulceration, gastritis, oesophagitis, liver disease (including but nol limited to ci ⁇ hosis, hepatitis).
  • These diseases or conditions may also include steroid-resistant diseases or conditions where treatment with a glucocorticoid is indicated, bat where the glucocorticoid is ineffective or is not as effective as expected.
  • the methods of the invention are preferably performed in a steroid-sparing manner.
  • steroid-sparing refers to a combination therapy method that allows a reduction in the amount of glucocorticoid administered while still providing an effective therapy for the disease or condition being treated or prevented.
  • Steroid-resistant diseases or conditions are diseases or conditions for which treatment with a glucocorticoid is indicated, but where the glucocorticoid is ineffective or is not as effective as expected. This term encompasses diseases or conditions for which the effective dose of glucocorticoid results in unacceptable side effects and/or toxicity. Some steroid-resistant diseases or conditions may require a dosage of glucocorticoid so large that they are considered non-responsive and therefore arc not ahlc to be successfully treated with glucocorticoids. Some steroid-resistant diseases or conditions may require a large dosage of glucocorticoid to achieve only a small effect on the symptoms of the disease or condition. Furthermore, some patients, diseases or conditions present with symptoms that do not respond to treatment with a glucocorticoid, or may become less sensitive to glucocorticoid treatment over time.
  • Glucocorticoids are a group of steroid hormones, which are used to treat or prevent a wide range of diseases or conditions. Suitable glucocorticoids may be synthetic or naturally occurring and include but are not limited to prednisolone, prednisone, cortisone acetate, beclamethasone, fluticasone, hydrocortisone, dexamethasone, methyl prednisolone, triamcinolone, budeso ⁇ ide and betamethasone.
  • the glucocorticoid used is selected from prednisone, prednisolone, hydrocortisone, fluticasone, betamethasone, betamethasone, methyl prednisolone, budesonide, triamcinolone, dexainelhasone and cortisone.
  • the glucocorticoid is selected from prednisone, prednisolone, methyl prednisolone, fluticasone and beclaincthasone, Beclameth&sone and fluticasone are particularly preferred for treating asthma.
  • Prednisone, prednisolone and methyl prednisolone are particularly preferred in the treatment of systemic or local inflammatory diseases.
  • the amounts of glucocorticoid and compound of formula (I) are selected such that in combination they provide complete or partial treatment or prophylaxis of a disease or condition for which a glucocorticoid is indicated.
  • the amount of compound formula (I) Ls preferably an amount that will at least partially inhibit the cytokine or biological activity of MIF.
  • the amount of glucocorticoid is preferably less than the amount required in the absence of the compound of formula (I).
  • the amounts of glucocorticoid and compound of formula (I) used in a treatment or therapy are selected such that in combination they at least partially attain the desired therapeutic effect, or delay onset of, or inhibit the progression of, or halt or partially or fully reverse the onset or progression of the disease or condition being treated.
  • the amounts of glucocorticoid and compound of formula (I) used in the prophylaxis of a disease or condition arc selected such that in combination they at least partially prevent or delay the onset of the .disease or condition. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods.
  • Suitable doses of a compound of formula (I) may lie within the range of about 0.1 ⁇ g per kg of body weight to 1 g per kg of body weighr per dosage.
  • the dosage is preferably in the range of 1 ⁇ g to 1 g per kg of body weight per dosage, such as is in the range of 1 mg to 1 g per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 500 mg per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 250 mg per kg of body weight per dosage.
  • the dosage is in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per kg of body weight per dosage, In yet another embodiment, the dosage is in the range of l ⁇ g to lrng per kg of body weight per dosage.
  • Suitable dosage amounts of glucocorticoids will depend, in part, on lhe mode of administration and whether the dosage is being administered in a single, daily or divided dose, or as a continuous infusion.
  • dosages When administered orally, intravenously, intramuscularly, intralesionally or intracavity (eg. intra-articular, intrathecal, intrathoracic), dosages are typically between 1 mg to lOOO nig, preferably 1 mg to 100 mg, more preferably 1 mg to 50 mg or 1 mg to 10 mg per dose.
  • dosages When administered topically or by inhalation as a single, daily or divided dose, dosages are typically 1 ng to 1 ⁇ g, 1 ng to 1 mg o ⁇ 1 pg to 1 ⁇ g.
  • Suitable dosage amounts and dosing regimens cau be determined by the attending physician or veterinarian and may depend on the desired level of inhibiting activity, the particular condition being treated, the severity of the condition as well as the general age, healLh and weight of the subject.
  • the glucocorticoid and compound of formula (I) ' may be administered simultaneously or sequentially.
  • the active ingredients may be administered alone but are preferably administered as a pharmaceutically acceptable composition or separate pharmaceutically acceptable compositions.
  • compositions of formula (I) are well known to those skilled in lhe art and are described above in relation to compounds of formula (I).
  • the composition or compositions may contain pharmaceutically acceptable additives such as carriers, diluents or excipients. These include, where appropriate, all conventional solvents, dispersion agents, fillers, solid carriers, coating agents, antifungal and antibacterial agents, dermal penetration agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
  • Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the glucocorticoids and/or
  • the compounds of formula (I), either as the only active agent oc together with another active agent, eg; a glucocorticoid, may also be presented for use in veterinary compositions. These may be prepared by any suitable means known in the ' ait. Examples of such compositions include those adapted for:
  • oral administration eg drenches including aqueous and non-aqueous solutions or suspensions
  • external application eg drenches including aqueous and non-aqueous solutions or suspensions
  • tablets boluses, powders, granules, pellets for admixture with feedstui ⁇ s, pastes for application to the tongue;
  • parenteral administration eg subcutaneous, intramuscular or intravenous injection as a sterile solution or suspension
  • topical application eg creams, ointments, gels. lotioiLS, etc.
  • compounds of Formula (I) or salts or derivatives thereof may be used us laboratory or diagnostic or in vivo imaging reagents. Typically, for such use the compounds would be labelled in some way, for example, radio isotope, fluorescence or colorknetric labelling, or be chelator conjugated.
  • compounds of Formula (!) could be used as part Of an assay system for MIF or as controls in screens for identifying other inhibitors. Those skilled in the art are familiar with such screens and could readily establish such screens using compounds of Formula (I). Those skilled in the art will also be familiar with the use of chelate conjugated molecules for in vivo diagnostic imaging.
  • Inhibitors of MlF may also b& used in implantable devices such as stents. Accordingly, in a further aspect the present invention provides an implantable device, preferably a stent, comprising:
  • the method is for inhibiting the cytokine or biological activity of MIF in a local region of the subject and the device is implanted within or proximate to die local region of the subject.
  • the present invention provides a method of treating, preventing or diagnosing a disease or condition wherein MIF cytokine activity is implicated comprising the step of implanting an implantable device according to the invention in a subject in need thereof.
  • the disease or condition is confined to a local region of the subject and the device is implanted with in or proximate to the local region.
  • the present invention further provides an angioplasty stent, for inhibiting the onset of restenosis, which comprises an a ⁇ gioplastic stent Qperably coated with a prophylactically effeclive dose of a composition comprising at least one compound of formula (T).
  • a ⁇ gioplastic stents also known by other terms such as “intravascular stents” or .simply “stents”, are well known in the art. They arc routinely used to prevent vascular closure due to physical anomalies such as unwanted inward growth of vascular tissue due to surgical trauma. They often have a tubular, expanding lattice- type structure appropriate for their function, and can optionally be biodegradable.
  • the stent can be operably coated with at least one compound or formula (I) using any suitable means known in the art.
  • "operably coating" a stent means coating it in a way that permits the timely release of the compott ⁇ d(s) of formula ( I) into the surrounding tissue to be treated once the coated stent is administered.
  • Such coating methods can use the polymer polypyrrole.
  • the present invention farther provides a method for inhibiting the onset of restenosis in a subject undergoing angioplasty, which comprises topically administering a stent according to the present invention to the subject at around the time of the angioplasty.
  • administration at around the time of angioplasty can be performed during the procedure, or immediately before or after the procedure.
  • the administering can be performed according to known methods such as catheter delivery.
  • stents that, release or elutc a pharmaceutical active
  • the standard approach is to use current highly refined metallic stent designs with polymer materials that release the active in a controlled manner.
  • polymer materials have been used for the coaling of stents to permit the eiution of drugs. These include bioerodible polymers such as poly-L lactic acid, biostable polymers such as polyurcthanc derivatives and slilicone-based polymers, as well as hydrogels.
  • a drug-eluting stent requires the drug io bo bound to ihe stent or its polymer or other coating in such a way as to allow steady release of drug over a period of time, and that the drug is able to be locally absorbed into cells in the vessel and stent lumen.
  • the optimum stent coating material and delivery parameters vary according to the tissue retention of the drug, such that rapid release of a tissue-retained drug can have long lasting effects, whereas a drug retained in tissues for a shorter time would need to be released over a longer period.
  • a person skilled in the art would be able to select, appropriate materials and conformations of stent for a particular purpose and particular small molecule inhibitor.
  • compounds of Formula (I) may be prepared using the methods depicted or described herein or known in the art. It. will be understood that minor modifications to methods described herein or known in the art may be required to synthesize particular compounds of Formula (I). General synthetic procedures applicable to the synthesis of compounds may be found in standard references such as Comprehensive Organic
  • reaction mixture was cooled, poured onio ice/water (500 nil) and filtered to give 5-broinoaeetyl ⁇ xi ⁇ dole as a light brown solid (7.1 g, 82%) that was used without further purification.
  • examples 14-21 were prepared by reaction of either 5- chloroaceiyloxuidole, 5-chlon>acetyl-6-chloroox indole, 6-chloroacetyl-2-beiizoxazolinone or ⁇ -bromoacetyl-Z-benzothiazoIinone, with 3-inercapto ⁇ ropioftie acid, 6-inercapto-l - hexanol, 1-b ⁇ ta ⁇ ethiol or thioglycolic acid.
  • the aqueous phase was extracted with ethyl acetate (75 ml) and the combined ethyl acetate extracts were washed with water (2 x 75 ml) and brine (75 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated to appro* 30 ml and chilled overnight. The mixture was then filtered and the residue dried under vacuum to give the title compound ( 1.429 g, 76% yield) as a brown powder, mp 211-213 0 C.
  • Aluminium chloride (13.427 g, 101 mmol) was suspended in DCE (80 ml), cooled in an ice bath and chloroacctyl chloride (6.4 ml, 80 mmol) added dropwi.se with a glass dropping pipette.
  • the mixture was stirred at O 0 C under nitrogen for 30min then 1 ,3-diinethyl- 1 ,3- dihydro-2tf- mecanicrnidazol-2-c> ⁇ e (6.504 g, 40-1 mmol) was added in portions.
  • the mixture was healed at 55 0 C under nitrogen for 2h, then allowed to cool to room temperature and poured onto ice (200 g). The mixture was filtered and the residue washed with water (100 ml).
  • the filtrate contained two phases which were separated. An attempt was made to dissolve the residue in a mixture of the DCE phase from the filtrate, additional DCH (50 ml) and chloroform (150 ml). The residue only partially dissolved and washing this mixture with water ( I (X) ml) gave an emulsion. The mixture in the separating funnel was filtered and the remaining solid in the separating funnel was suspended in water (3 x 100 ml) and ethyl acetate (40 ml). Each of the washes was filtered and the residue was dried at the pump and then dried under vacuum over silica gel overnight to give the title compound (7.003 g, 85% yield) as a pink solid.
  • the aqueous phase was extracted with ethyl acetate (60 ml) and the combined ethyl acetate extracts were washed with water (2 X 60 ml) and brine (60 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give an orange oil which solidified on standing. The solid was broken up to give the title compound (1.868 g, 95% yield) as a yellow powder, mp 80-81 0 C.
  • Aluminium chloride (9.953 g, 74.6 mmol) was suspended in DCE (20 ml) and cooled in an ice bath. Chloroacetyl chloride (4.70 nil, 59.0 mmol) was added dropwise with a glass dropping pipette and the mixture was stiired at O 0 C under nitrogen for 30min. 5-Chloro- l,3-dihydro-2/J-bcnzimidazol-2-one (5.0(K) g, 29.7 mmol) was added in portions and the mixture was heated at 55"C under nitrogen for 3Vih. The mixture was allowed to stand at room temperature under nitrogen overnight, lhen heated at 55°C under nitrogen for a further 5VJII.
  • the reaction mixture was allowed to cool Io room temperature and poured onto ice (400 g) and filtered. The residue was washed with water (2 x 100 ml) and dried at the pump. The residue was washed with ethyl acetate (20 ml, 3 x 40ml) and dried at the pump to give the title compound (2.631 g, 36% yield) as a dark green powder.
  • the product contained 10 mol% 5-chloro-l,3-dihydro-2H-bcnzimidazol-2-o ⁇ e.
  • the aqueous pliase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (50 ml). A further portion of ethyl acetate (100 ml) was added to lhe ethyl acetate phase and the ethyl acetate extracts were washed with water (50 ml) and brine (50 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give a red grey powder (1.441 g, 91% yield). 1 H nmr analysis showed that the product contained 10 mol% unchanged starting material.
  • 3-Mercapto ⁇ ropionic acid (566 nig, 5.33 minol) was dissolved in anhydrous DMH" (17 ml) and 5-chloro-6 ⁇ (chloroacctyl)-13-dihydra-2H-bcn7.imidazol-2-one (1.30U g, 5.30 nunol) and anhydrous potassium carbonate (3.714 g, 26.9 itunol) were added.
  • the a ⁇ xture was stirred under nitrogen for 35min then the reaction mixture was partitioned between ethyl acetate (100 ml) and water (150 ml). Emulsions prevented the separation of the phases and hydrochloric acid (J M 1 80 ml) was carefully added.
  • the phases were separated and the aqueous-phase was extracted with ethyl acetate (100 ml, 50 ml).
  • the combined ethyl acetate extracts were washed with water (2 x 100 ml) and brine (100 ml), dried over anhydrous magnesium sulfate, and filtered.
  • the filtfaic was evaporated to dryness to give a dark green powder.
  • the crude product was partitioned between ethyl acetate (150 ml) and a 5% sodium hydrogen carbonate solution (200 ml).
  • the ethyl acetate phase was extracted with water (100 ml) and the combined aqueous phases were washed with ethyl acetate (100 ml), acidified with hydrochloric acid (3M, 50 ml) and extracted with ethyl acetate (300 ml, 2 x 100 rnl). There was a significant quantity of a pale brown solid that did not dissolve.
  • the aqueous phase containing the emulsion was filtered and dried at the pump to give the title compound (447 mg, 27% yield) as a cream solid.
  • the ethyl acetate extracts were evaporated to dryness to give a green solid and the residue partitioned between ethyl acetate (ISO ml) and a 5% sodium hydrogen carbonate solution (200 ml).
  • the aqueous phase was acidified with hyd ⁇ oclilorie acid (3M, 50 ml) and the resultant suspension washed with ethyl acetate (50 ml).
  • the combined aqueous and ethyl acetate phases were filtered and the residue was washed with water (2 x 50 ml) and dried at the pump to give the title compound (579 mg, 35% yield) as a pale green solid.
  • Aluminium chloride (8.589 g, 64.4 mmol) was suspended in DCE (17 nil) arid cooled in an ice bath. Chloroacetyl chloride (4.05 ml, 50.8 mmol) was added dropwise with a glass dropping pipette and the mixture was slirred at O 0 C under nitrogen for 30min. 5-Chloro- l,3-dimethyl-l,3-dihydro-2J/-benzimidazol-2-one (5.010 g, 25.5 mmol) was added in portions and Lhc mixture was heated at 55°C under nitrogen for 3h. The mixture was allowed to cool to room temperature and poured onto ice (200 g) then filtered. The residue was washed with water (3 x 100 ml), dried at the pump and then dried under vacuum over silica gel to give the title compound (5.573 g, 80% yield) as a brown powder.
  • the aqueous phase was extracted with eihyl acetate (60 ml) and the ethyl acetate extracts were washed with water (2 x 60 ml) and brine (60 ml), dried over anhydrous magnesium sulfate and filtered.
  • the filtrate was evaporated to dryness and the resultant residue purified by bulb-to-bulb distillation (250 0 C / 0.57mbar) to give the title compound (1.037 g, 54% yield) as a pale yellow solid, mp 66-68 0 C,
  • 3-Mcrcaptopropio ⁇ ie acid (593 mg, 5.59 mmol) was dissolved in anhydrous DMF (17 ml) and 5-chloro-6-(chloi'oacetyl)-l,3-dimethyl-l,3-dihydro-2W-ben7.imida2ol-2-onc (1.498 g, 5.48 minol) and anhydrous potassium carbonate (3.784 g, 27.4 mmol) were added. The mixture was stirred under nitrogen for 35min then partitioned between ethyl acetate (100 nil) and hydrochloric acid ( 1 M, 80 ml).
  • the aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 50 ml) and brine (50 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (1.797 g, 96% yield) as a bruwn powder, mp 148-150 0 C.
  • the aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 100 ml) and extracted with a 5% sodium hydrogen carbonate solution (200 ml) and water (50 ml). These extracts were acidified with hydrochloric acid (3 M, 50 ml) and filtered and the residue was dried under vacuum over silica gel to give the title compound (1.105 g, 63% yield based on available bromoacetyl and chloroacetyl compounds) as a pale yellow solid, mp 195-197 0 C.
  • 3-Mercaptopropionic acid (515 mg, 4.85 mmol) was dissolved in anhydrous DMF (15 ml) and 6-(bromoacely ] )-5-chloro-3-methyl-l,3-bcnzothiazol-2(3H)-one (1.551 g, 4.84 mmol) and anhydrous potassium carbonate (3,595 g, 26.0 mmol) were added.
  • the mixture was siirced under nitrogen for 40 mill then the reaction mixture was partitioned between ethyl acetate (150 ml) and hydrochloric acid (IM, 80 ml).
  • the aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 100 ml), and extracted with a 5% sodium hydrogen carbonate solution (200 ml), and water (50 ml). These extracts were acidified with hydrochloric acid (3M, 50 ml) causing a brown oil to precipitate.
  • the mixture was extracted with ethyl acetate (100 ml, 50 ml) and the ethyl acetate extracts were washed with brine (75 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (1.476 g » 88% yield) as .a cream powder, mp 120-121 0 C.
  • Example 2 In vitro assay of MIF antagonism: inhibition of LPS-i ⁇ duced production of IL-6 in RAW264.7 macrophages by compounds
  • MIF is an important factor in the innate immune response to toxins such as the bacterial endotoxin lipopolysaccharide (LPS).
  • LPS bacterial endotoxin lipopolysaccharide
  • endogenous M (F activity is required for expression of the LPS receptor toll-like receptor-4 ⁇ 12 I
  • a compound with the ability to inhibit the biological activity of MlF would therefore inhibit the activation of cytokine production by macrophages in response to LPS.
  • the RAYV264.7 mouse macrophage cell line was propagated in DMEM/l 0% foetal calf serum (FCS) a. 37 0 C in 5% CO 2 . 24 hr prior to assay cells were seeded in 96-wcll tissue culture plates. Cells were allowed to adhere for 4 hi prior to transfer to DMEM/0.5% FCS for 18 hr. Cells were then treated with 50 uM compound in DMSO for 30 min prior to stimulation for 4 hr with 100 ng/ml LPS. Cell culture supcm ' atants were then collected from each well and assayed for IL-6 levels by ELISA (R&D Systems) according to the manufacturer's Instructions.
  • FCS foetal calf serum
  • Figure 1 shows that Compound 19 treatment induces a dose-dependent inhibition of LPS- induced IL-6 production when RAW264.7 cells are pre-ireated with up to 100 ⁇ M concentration of compound and the samples analysed for JL-6 production as described above.
  • the IC50 value for the compound was determined to be 20 uM.
  • Table 2 shows the % inhibition of IL-6 production induced by 50 uM compound treatment relative to LPS + PMSO control levels (with basal levels of IL-6 in llic absence of LPS subtracted). The compounds induce marked decreases in IL-6 production consistent with antagonism of endogenous MlF. Table 2. Inhibition of LPS-induced IL-6 production in RAW264.7 cells
  • Example 3 In vitro assay of MIF antagonism: inhibition of interlqukin-l induction of cycloxygenase-2 expression in S 112 human dermal fibroblasts by Compounds
  • Sl 12 human dermal fibroblasts were propagated in RPMI/10% foetal calf serum (FCS). Prior to experimentation, cells were seeded at 10 5 cells/ml in RPMI/0.1% BSA for 18 hours. Cells were treated with recombinant human IL-I (0.1 ng/ml) and with each compound, at concentrations raingi ⁇ g up to 100 ⁇ M. A control was treated only with recombinant human IL-I (0.1 ng/ml) and vehicle (DMSO). After 6 hours, cells were collected and intracellular COX-2 protein determined by pcrmeabilisation flow cytometry.
  • Cells permeabilised with 0.1% saponin were sequentially labelled with a mouse anti-human COX-2 monoclonal antibody and with shecp-anti-mouse F(ab)2 fragment labelled with fluorosccin isothioeyanate.
  • Cellular fluorescence was determined using a flow cytometer. At least 5000 events were counted for each reading, each of which was performed hi duplicate, and the results expressed in mean fluorescence intensity (MFI) after subtraction of negative co ⁇ trol- labellcd cell fluorescence.
  • MFI mean fluorescence intensity
  • Figure 2 shows treatment with Compound 2 induces a dose-dependent inhibition of IL-I induced COX-2 expression when S 1 12 cells are treated with up to 100 ⁇ M concentration of compound and the samples analysed for COX2 expression as above. The results show significant and dose-dependent reductions in COX2 expression levels consistent with antagonism of M IF activity.
  • Endotoxaemia was induced by intra-perito ⁇ eal Injection of C57B1/6J mice with lipopolysaccharidc (LPS) (lmg/kg) in 200 ⁇ l saline.
  • Animals were treated with either a saline solution (control) only, or LPS with vehicle or compounds Bl and A3 in vehicle at doses of 10, 1 and 0. t mg ⁇ tg body weighL, administered by intra-peritoncal injection at 24 hours and 1 hour before intra-peritoneal LPS injection. After I hour mice were humanely killed by CO2 inhalation then neck dislocation. Serum was obtained from blood obtained by cardiac puncture prior to death and measured for TNF levels by ELISA according to the manufacturer's instructions.
  • LPS lipopolysaccharidc
  • MlF protein has the ability In vitro to catalyze the tautisomerizalion of dopachrome ⁇ 15 ⁇
  • the tautomerase activity of MIH is unique, as is the structure and sequence of the section of MIF responsible for this phenomenon, suggesting that small molecules binding to or docking in this site would be specific for MIF.
  • the relevance of this enzymatic activity to the development of inhibitors of the cytokine and biological activity of MIF is that demonstration of inhibition of taulison ⁇ er ⁇ se activity is a demonstration that a given compound has a direct physical interaction with M ⁇ F.
  • Delayed-type hypersensitivity reactions which are itiitatcd by T lymphocyte responses to recall antigens and mediated by many cell types including macrophages, are known to be dependent on the cytokine or biological activity of MIF (16 ' I7) .
  • an anti-MIF monoclonal antibody suppresses delaycd-typc hypersensitivity reactions reactions in vivo to methylated bovine scrum albumin (mBSA) injected inio the skin of animals preimmunised with mBS ⁇ 06 K
  • mBSA methylated bovine scrum albumin
  • a compound inhibiting Lhc cytokine or biological function of MIF might be expected to inhibit delayed -type hypersensitivity reactions in viv ⁇ .
  • mice were immunised on day 0 with 200 ⁇ g of methylated BSA (niBSA;.Sigma Chemical Co., Castle Hill, Australia) emulsified in 0.2 ml of Freu ⁇ d's complete adjuvant (CFA; Sigma) injected subculaneously in the flank skin.
  • CFA Freu ⁇ d's complete adjuvant
  • mice were given 100 ⁇ g tnBSA in 0.1 ml CFA by intradermal injection at the base of the tail.
  • Mice were challenged on day 27 following first immunisation by a single intradermal (ID) injection of 50 ⁇ g mBS ⁇ /20 ⁇ l saline in the right footpad, with 20 ⁇ l saline injected in the left footpad serving as control (Santos, 2001).
  • ID intradermal
  • mice were killed 24 h later and footpad swell ing quantified using micro calipers (Mitutoyo, Kawasaki-shi, Japan). DTH measurements were performed by an observer blinded to mouse genotype. Results were expressed as the difference in footpad swelling between ⁇ iBSA and saline-injected footpads, and expressed as change in footpad thickness (mm). Mice were treated with compound 13 at 5 and 15 mg/kg/24h by EP injection, twice daily for 7 days prior to antigen challenge with rnBSA in the footpad. Treatment with compound 13 continued for a further 24h and changes in footpad thickness relative to control paws were measured at that time. As shown in Figure 4, compound 13 induced a significant inhibition of DTH reactions.
  • MIF is implicated in lhe recruitment of leukocytes to sites of inflamma ⁇ on, via studies which show that MIF-deficient mice exhibit reduced interactions between leukocytes and vascular endothelium in vzW ls> . More recently, it has been demonstrated that the administration of MIF in vivo induces the recruitment of macrophages to tissue (l9 ⁇ a process which first requires the induction of adherence o ⁇ circulating leukocytes to the vascular endothelial cells. As will be known to those skilled in the art, the adherence of leukocytes to the endothelium in vivo can be sLudied using the technique of intravital microscopy (l8 - 19) .
  • MIF induces leukocyte adherence to vascular endothelium as measured using intravital microscopy
  • a compound inhibiting the Cytokine or biological activity of MIF might be expected to inhibit the effects of MIF observable using intravital microscopy.
  • mice were anesthetised with kelamine/xylazinc, and the crcmastcr muscle was exteriorized onto an optically-clear viewing pedestal.
  • the cremasteric microcirculation was visualized using an intravital microscope (Axioplan 2 Imaging; Carl Zeiss, Australia) with a 2OX objective lens (LD Achroplaii 20X/0.40 NA, Carl Zeiss, Australia) and a 1OX eyepiece. Three-five postcapillary venules (25-40 ⁇ m in diameter) were examined for each experiment. Images were visualized using a video camera and recorded on video-tape for subsequent playback analysis.
  • Recombinant human MTF (1 mg) was injected inlrascrolally in 150 ⁇ L saline, prior to intravital microscopy 4 hours later.
  • Leukocyte-endotheUal cell adhesion was assessed as described by Gregory et al (l9 ⁇ Compound 13 at a dose of 30 mg/kg or vehicle were administered by intraperitoneal injection 10 minutes prior to intrascroial injection of MIF.
  • MIF induced leukocyte adhesion markedly above baseline leukocyte adhesion observed without MIF injection (dotted line).
  • MlF-induced leukocyte adhesion was reduced approximately 50 % by compound 13 administration.
  • aqueous solubility of the compound is sufficiently high to allow dosing of humans with a pharmacologically active dose.
  • Compounds with only limited aqueous solubility may be less suitable for development as a human therapeutic.
  • aqueous compound solubility was determined in a nepholometer in phosphate-buffered saline containing ⁇ .005% (v/v) P20 and a. final concentration of 5% DMSO. Briefly, compounds were initially dissolved in DMSO as a 10 mM stock solution and diluted lo 1 mM. and 0.5mM working solutions with neat DMSp. The compounds were then titrated in DMSO and a constant volume of DMSO stock added to filtered PBS/P20 solution so that the final DMSO concentration was 5%. The solubility was then determined in clear, flat-bottom 96- well plates using the nephclometer and reported as the concentration range at which the compound begins to precipitate from solution. Results
  • MlF is a pituitary-derived cytokine . that potentiates lethal endotoxaemia. Nature 365: 756.
  • MIF macrophage migration inhibitory factor

Abstract

The present invention relates to the use of specific benzimidazolone analogues and derivatives to inhibit the cytokine or biological activity of macrophage migration inhibitory factor (MIF), and diseases or conditions wherein MIF cytokine or biological activity is implicated. Novel benzimidazole analogues and derivatives are also provided.

Description

MIF INHIBITORS
FIFXD OF THE I-WENTION
The present invention relates generally to the treatment of diseases or conditions resulting from cellular activation, such as inflammatory or cancerous diseases or conditions. In particular, the invention relates to the use of specific bcnzimidazolone analogues and derivatives to inhibit the cytokine or biological activity of macrophage migration inhibitory factor (MIF), and diseases or conditions wherein MIF cytokine or biological activity is implicated.
BACKGROUND OF THE INVENTION
MIF is the first identified T-cell-derived soluble lymphokiαe. MIF was first described as a soluble factor with the ability to modify the migration of macrophages ^1K The molecule responsible for the biological actions ascribed to MIF was identified and 'cloned in 1989 (2l Initially found to activate macrophages at inflammatoiy sites, it has been shown to possess pluripøtential actions in the immune system. MIF has been shown to be expressed in human diseases which include inflammation, injury, ischaeroia or malignancy. MIF also has a unique relationship with glucocorticoids by overriding their and- inflammatory effects.
Recent studies have indicated thai monoclonal antibody antagonism of MIF may be useful in the treatment of sepsis, certain types of cancers and delayed type hypersensitivity. Antibody antagonism of MIF has also been shown to have activity in adjuvant- or collagen-induced arthritis animal models and models of other inflammatory and immune diseases including colitis, multiple sclerosis, atherosclerosis, glomerulonephritis, and uveitis.
Although antibody antagonism of MIF i& one potential way to provide therapeutic treatments, such biological molecules can be expensive to prepare on a commercial basis and further, can be limited in the way they are administered (generally by injection) arid do not readily lend themselves to formulations for administration by other means eg oral administration. Small molecule inhibitors may overcome one or more such difficulties connected with the use of biological therapeutic treatments. There exists a need, therefore, for small molecule inhibitors of the cytokine or biological activity of MIF. Small molecule inhibitors of the cytokine or biological activity of MIF would have, therapeutic effects in a broad range of diseases, whether given alone or in combination with other therapies.
Further, glucocorticoids have been used to treat human diseases for over fifty years and are effective in ιχ range of diseases which include inflammation, injury, ischacmia or malignancy. Although debate continues in relation to their impact on disease progression, their influence on symptoms and signs of inflammation, especially in the short term, can be dramatic.
Despite their benefits and efficacy, the use of glucocorticoids is limited by universal, predictable, dose- dependent toxicity. Mimicking Cushing's disease, a disease wherein the adrenal glands produce excess endogenous glucocorticoids, glucocorticoid treatment is associated with side effects including immunosuppression (resulting in increased susceptibility to infections), weight gain, change in body habitus, hypertension, oedema, diabetes mellitus, cataracts, osteoporosis, poor wound healing, thinning of the skin, vascular fragility, hirsutism and other features of masculinization (in Females), In children, growth retardation is also noted. These side effects are known as Cushiiigoid side effects.
Since the side effects of glucocorticoids are dose dependent, attempts to reduce the dosage requirement have been investigated, including combination therapies in which glucocorticoids are administered with other therapeutic agents. These combination therapies arc sometimes referred to as "steroid-sparing" therapies. However, currently available combination therapies are non-specific as the other therapeutic agents do not address biological events which inhibit the effectiveness of glucocorticoids. Such combination therapies are also typically associated with serious side effects.
Furthermore, glucocorticoids arc incompletely effective in a number of disease settings, leading to the concept of "steroid-resistant" diseases. Agents which amplify or enhance the effects of glucocorticoids would not only allow the reduction of dose of these agents but may also potentially fender "steroid-resistant" diseases steroid-sensitive.
There is a need for effective therapies which enable a reduction in the dosage level of , glucocorticoids. There is also a need for effective treatment of "steroid-resistant" diseases. Preferably, such therapies or treatments would address facLors which directly limit the effectiveness of glucocorticoids.
Therapeutic antagonism of MTF may provide "steroid -sparing" effects or be therapeutic in "steroid-resistant" diseases. Unlike other pro-inflammatory molecules, such as cytokines, the expression and/or release of M IF can be induced by glucocorticoids ^' <4\ Moreover, MlF is able to directly antagonize the effects of glucocorticoids. This has been shown to be the case for macrophage TNF, IL-I β, (L-6 and 1L-8 secretion (5X (6>, and for T cell proliferation and IL-2 release (7). In vivo, MlF exerts a powerful glucocorticoid-atitagonist effect in models including eπdotoxic shock and experimental arthritis fs)l ^K fn the context of an inflammatory or other disease treated with glucocorticoids, then, MIF is expressed but exerts an effect which prevents the glucocorticoid inhibition of inflammation. It can therefore be proposed that therapeutic antagonism of MIF would remove MIF' s role in inhibiting the anti-inflammatory effect of glucocorticoids, thereby allowing glucocorticoids to prevail. This would be the first example of true "steroid-sparing" therapy. In support of this hypothesis is the observation that anti-MIF antibody therapy reverses the effect of adrenalectomy in rat adjuvant arthritis ®K In further support of this, it has recently been demonstrated that reduced MIF activity is indeed directly associated with improvements in responsiveness to glucocorticoids (i(>'2l). By neutralizing the natural glucocorticoid 'counter-regulator' effect of MIF, it is envisioned that with MIF antagonism, steroid dosages could be reduced or even eliminated in inflammatory disease, particularly in those diseases that are associated with the glucocorticoid resistance (10)> {l [\ There is a need, therefore, for therapeutic antagonists of the cytokine or biological activity of MIF.
MIF has recently been shown to be important in the control of ieukocytc-cndothelial interactions. Leukocytes interact with vascular endothelial cells in order to gain egress from the vasculature into tissues. The role of MIF in this process has been demonstrated to affect in particular lcukocytc-endotlieliiil adhesion and emigration (22έ 23). This process is an essential part of nearly all inflammatory diseases, and also for diseases less well-identified as inflammatory including atherosclerosis (24). There is a need, iherefore, for antagonists of MTF to limit the recruitment of leukocytes into inflammatory lesions and lesions of diseases such as atlieroseleTosis.
In WO 03/104203, the present applicant has shown that certain benzimidazolc derivatives are capable of acting as inhibitors of MIF. The present inventors have now found a novel class of MlF inhibitors, members of which show impτoved characteristics as drug-like molecules when compared to the compounds of the prior art.
SUMMARY OF THE INVENTION
In a first aspect, the present invention provides a method of treating, diagnosing or preventing autoimmune diseases, tumours, or chronic or acute inflammatory diseases comprising administering a treatment, prevention or diagnostic effective amount of a. compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof wherein:
Figure imgf000005_0001
X is selected from- -O-, -S-, -C(R5)(Ry)- and -N(Rn)-;
Y is selected from - -N(R7)-, -O-, -S-, and -C(R7J2-; R, is selected from hydrogen, Ci-Q,alkyl,
Figure imgf000006_0001
C(RsRsOi1SR7. (CRsRs1X1N(TIs)2 and (CR5R5-X4TIaIo;
R3 is selected from hydrogen, C,-Cfialkyl, (CR16R16OpNR14R is, (CR1<sRι<v)rOR,7, (CR16R1 6OpSR171 (CR|6R1ff)phalo, (CR16R 1C')PNO2, (CRR1 ft.)πC(O)R28, (CR10R16OnS(O)R17, (CR10R16OnS(O)2R17, (CR10RInF)nS(O)3R17. and
Figure imgf000006_0002
R4 is selected from hydrogen, halogen, Ci-Csalkyl, C^-Caalkenyl, C2-C:salkynyl and
Figure imgf000006_0003
each Rs and R.y is independently selected from hydrogen, C]-Q>alkyl, halo, OR7, SR7 and N(Rn)2;
each Rn is independently selected from hydrogen, Ci-Csalkyl and 0Ii7;
each R7 is independently selected from hydrogen and Ci-C3alkyl;
each R1.2 and R 12' is independently selected from hydrogen, Ci-Csalkyl, C2-C6alkenyl, C2- Cήalkynyl, OR24, SR24, halo, N(R24)Z, CO2R24, CN, NO2, aryl and heteracyclyl;
each RM and Rn is independently selected from hydrogen,
Figure imgf000006_0004
OR]7, SR17, and
each R1ri and R1^ is independently selected from hydrogen, CrQalkyl, halo, OR]7, SR]7 and N(RI7)2;
each R)7 is independently selected from hydrogen and Ci-Csalkyl;
each R1g is independently selected from hydrogen and halo;
R22 is selected from Ci-Qalkyl, NH2, NH(Cι-C6alkyl), N(Ci-CcaIkyl)2, OR29 or SR29;
each R24 is selected from H and Ci-Cgalkyl; R.28 is selected from hydrogen, Ci-C6aLkyl, OR29, SR29 or N(R2P)2;
each R20 is independently selected from hydrogen and
Figure imgf000007_0001
Q is selected from O, S, NR40, S(O)11 where u is an integer from ϊ to 2;
R4Q is selected from H, OH, and C(R4IR1UOvR4Z;
each R41 and R4ι< is independently selected from H, OH, halo, NH2, cyaπo, and NO2;
R42 is independently selected from H1 OR43, COOR43, CON(R43^1), O(CO)R43, aryl, and heterocyclyl;
each R43 and Ro- is independently selected from H, Cj oalkyl, benzyl, and aryl;
a = O or an integer to 3
m is 0 or an integer from 1 to 20;
p is 0 or an integer from 1 to 6;
I is an integer from 1 toll)
v is 0 or an integer from i to 10.
In particular, the autoimmune disease, tumour, or chronic or acute inflammatory disease is selected from the group comprising:
rheumatic diseases (including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis) spondyloarthropathies (including but not limited to ankylosing spondylitis, reactive arthritis, Reiler's syndrome), crystal arthropathies (including but not limited to gout, pscudogout, calcium pyrophosphate deposition disease), Lyme disease, polymyalgia rhcumatica;
connective tissue diseases (including but not limited to systemic lupus syndrome);
vasculitides (including but not. limited to polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome);
inflammatory conditions including consequences of trauma or ischaemia;
sarcoidosis;
vascular diseases including atlicrosclciOtic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischaemic heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis;
ocular diseases including uveitis, corneal disease, iritis, iridocyclitis, cataracts; autoimmune diseases (including but not limited to diabetes mcllitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis);
pulmonary diseases (including but not. limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome);
cancers whether primary or metastatic (including but not. limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
renal diseases including glomerulonephritis, interstitial nephritis;
disorders of the hypolhalamic-piluiiary-adrenal axis;
nervous system disorders including multiple sclerosis, Alzheimer's disease;
diseases characterised by modified aiigiogenesis (eg diabetic retinopathy, rheumatoid arthritis, cancer), endometrial function (menstruation, implantation. complications of infective disorders including endotoxic (septic) shock, cxotosic (septic) shock, infective (true septic) shock, malarial complications^ other complications of infection, pelvic inflammatory disease;
transplant rejection, graft-versus-host disease;
5. allergic diseases including allergies, atopic diseases, allergic rhinitis;
bone diseases (eg osteoporosis, Paget's disease);
skin diseases including psoriasis, atopic dermatitis, UV(B)-induced dermal cell activation (eg sunburn, skin cancer);
diabetes mellitus and its complications;
0 pain, testicular dysfunctions and wound healing;
gastrointestinal diseases including inflammatory bowel disease (including but not limited to ulcerative colitis, Crohn's disease), peptic ulceration, gastritis, oesophagitis, liver disease (including but not limited to cirrhosis, hepatitis).
MIF cytokine or biological activity is implicated in flic above diseases and conditions.
5 Preferably, die disease or condition is selected from, the group consisting of rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, multiple sclerosis, psoriasis, uveitis, diabetes mellitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and chronic obstructive pulmonary disease.
In a second aspect, the present invention provides a compound of Formula (II) or a 0 pharmaceutically acceptable salt or prodrug thereof wherein:
Figure imgf000010_0001
II
X is selected from - -O-, -S-, -C(R5)(R5-)- and -N(R6)-;
Y is selected from - -N(R7)-, -O-, and -S-;
Z is selected from >C=O, >C=S, and >C=NRd;
R1 is selected from hydrogen, Ci-C3alkyl, (CRsRsO11OR7, C(R5R5OnSR7, (CR5R5OI1N(RG)2 and (CRsR5')nhalo;
R1 is selected from hydrogen, CrCftalkyl, (CRi0R 10OpNR14RiS, (CR1eR1sOpORπ, (CR, 6R16OpSR17, (CR1gR1eOphalo, (CR16R1nOpNO2, (CRlftRIft.)nC(O)R2H, (CRlftR]ft)nS(O)R17, (CR16R16OaS(O)2R17, (CR16Ru)nS(O)SR17, and
Figure imgf000010_0002
R4 is selected from hydrogen, halogen, C|-C3alkyl, C2-C3alkenyl, Cz-Csalkynyl and
Figure imgf000010_0003
each R5 and R? is independently selected from hydrogen, Ci-Cjalkyl, halo, OR7, SR7 and N(Re)2;
each Rf, is independently selected from hydrogen, Ci-C3alkyl and OR7;
each R7 is independently selected from hydrogen and Ci-C^alkyl; each R12 and R12' is independently selected from hydrogen, C1-C(JaIlCyI, C2-Cήalkeπyl, C2- C6alkynyl, OR24, SR24, halo, N(R24^, CO2R24, CN, NO2, aryl and heterocyclyl;
each R14 and R15 is independently selected from hydrogen, CrC^alkyl, OR17, SR17, and N(Rn)2;
each R16 and R16- is independently selected from hydrogen, C1-C3alkyl, halo, ORn, SRn and N(R17)2;
each R [7 is independently selected from hydrogen and Ci-C^alkyl;
each R1S is independently selected from hydrogen and halo;
R22 is selected from Ci-C6alkyl, NH2, NH(Ci-C6aikyl), N(Ci-C^alkyl)2, OR29 or SR20;
each R24 is selected from H and Ci-Cβalkyl;
R28 is selected from hydrogen, Cj-Qalkyl, OR20, SR20 or N(R2(J)2;
each
Figure imgf000011_0001
Q is selected from O , S , S(0)u where u is an integer from 1 to 2;
R40 is selected from 117 OH, and C(R4IR41OvR42;
each R41 and R^' is independently selected from H, OH, halo, NH2, CN and NO2;
R42 is selected from H, OR4.1, COOR4:!, CON(Ri3R4S-), 0(CO)R43,
Figure imgf000011_0002
aryl, and hcterocyelyl;
each R43 and R«- is independently selected from 11, Cj 0 alky], and benzyl;
n is O or 1 to 3;
m is O or an integer from 1 to 8; p is ϋ or an integer from 1 to 6;
t is an integer from 1 to 10;
v is 0 or an integer from 1 to 10
provided that the compound is not
Figure imgf000012_0001
In a third aspect, the present invention provides a compound of Formula IH or a pharmaceutically acceptable salt or prodrug thereof wherein:
Figure imgf000012_0002
III
X is selected from - -O-, -S-, -C(R5)(Rs')- and -N(R6)-;
Y is selected from - -N(R7), -O-, and -S-; R1 is selected from hydrogen, Crdalkyl, (CR5Ks^OR7, C(R5R5OnSR7, (CR5R5OnN(Re)2 and (CR5R5')αhalo;
R;* is selected from hydrogen, C,-Cftalkyl, (CR10RK1O15NRIAR15, (CR16R16OpOR17, (CRwR16-)pSR17, (CR1βR1rfphalo, (CRJ6RIOOPNO2, (CR16R1 -OnC(O)R2S, (CR,«R,6.)πC(=NRM)RZ2, (CR16R1nOS(O)R177 (CR16R15OnS(O)2R17, (CR16R]6OaS(O)3R17, and
(CR|«R|6')pC(R]8)3;
R4 is selected from hydrogen, halogen, Cj-Qsalkyl, C2-C.ialkenyl, Cj-Cjalkynyl and (CR12Ri2On(CR1S)3; .
each R5 and Rj' is independently selected from hydrogen, C]-Cjalkylf halo, OR7, SR7 and N(Re)2;
each Rfi is independently selected from hydrogen, Cι-C3alkyl and OR7;
each Rγ is independently selected from hydrogen and Q-Cjalkyl;
each Rn and R12' is independently selected from hydrogen, Cj-Cftalkyl, C2-Cήalkenyl, C2- Cgalkynyl, OR24, SR24, halo, N(R^)2, CO2R24, CN, NO2, ary! and heterocyclyl;
each R14 and R15 are independently .selected from hydrogen, C[-C3alkyl, OR17, SR17, and N(R17)2;
each R16 and Rw is independently selected from hydrogen, Ci-Cjalkyl, halo, OR]7, SR17 and N(Rn)2;
each Rn is independently selected from hydrogen and Ci-C3alkyl;
each Rι« is independently selected from hydrogen and halo;
R22 is selected from Cj-Cβalkyl, NH2, NH(Ci -Cήalkyl), N(C]-C<;alJ<:yl)2, OR29 or SR20;
each R24 is selected from I ( and Ci-Cβalfcyl; R1 S is selected from hydrogen, Ci-Cealkyl, OR29, SK59 or N(R29)^;
each R29 is independently selected from hydrogen and Q-Caalkyl;
Ru in selected from OH, C(R45R45')vRύή;
each R45 and Rj5- is independently selected from H, OH, halo, NH2, CN, NO2;
each R46 is selected from COOR47, CON(R47R47'), 0(CO)R47, N(R47R4/);
each R47 and R47' is independently selected from H, C|.c alkyl» benzyl;
wherein when v is greater than 1 , R4^ can be OR47;
wherein when v is greater than 2 , R^n can be H;
n is O or 1 10 3;
m is O or an integer from 1 to 8;
p is O or an integer from 1 to 6;
t is an integer from 1 to 10;
v is O or an. integer from 1 to 10
provided that the compound is not
Figure imgf000014_0001
A further aspect of the invention provides for the use of a compound of Formula (I) pr a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a. medicament for the treatment of a disease or condition as above. A further aspect of the invention provides a pharmaceutical composition comprising a compound of the second or third aspect and a pharmaceutically acceptable carrier, diluent or excipient.
In a further aspect, the present invention provides a method of inhibiting cytokine or biological activity of MIF comprising contacting MIF with a cytokine or biological inhibiting amount of a compound of formula (I), or a pharmaceutically acceptable salt or prodrug thereof.
In another aspect, the invention provides a method of treating, preventing or diagnosing a disease or condition wherein MfF cytokine or biological activity is implicated comprising the administration of a treatment, prevention or diagnostic effective amount of a compound of formula (T) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof.
In a further aspect there is provided Qic use of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment, prevention or diagnosis of a disease or condition wherein MIF cytokine or biological activily is implicated.
In another aspect, the invention provides a method of treating or preventing a disease or condition wherein MIF cytokine or biological activity is implicated comprising:
administering to a mammal a compound of formula (I) and a second therapeutic agent.
In another aspect, the present invention provides a method of prophylaxis or treatment of a disease or condition for which treatment with a glucocorticoid is indicated, said method comprising:
administering to a mammal a glucocorticoid and a compound of formula (I).
In yet another aspect, the present invention provides a method of treating steroid-resistant administering to a mammal a glucocorticoid and a compound of formula (I).
In a further aspect, the present invention provides a method of enhancing the effect of a glucocorticoid in mammals comprising administering a compound of formula (I) simultaneously, separately or sequentially with said glucocorticoid.
In yet a further aspect, the present invention provides a pharmaceutical composition comprising a glucocorticoid and a compound of formula (I).
In a further aspect of the invention there is provided a use of a glucocorticoid in the manufacture of a medicament, for administration with a compound of formula (I) for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
In yet a further aspect of the invention there is provided a use of a compound of formula (I) in the manufacture of a medicament for administration with a glucocorticoid for the treatment or prophylaxis of a disease or condition for which treatment of a glucocorticoid is indicated.
In yet a further aspect of the invention there is provided a use of a glucocorticoid and a compound of formula (T) in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
Inhibitors of MIF may also be used in implantable devices such as stems. Accordingly, in a further aspect the present invention provides an implantable device, preferably a stent, comprising;
(i) a reservoir containing at least one compound of formula (I); and
(ii) means to release or elule the inhibitor from the reservoir
There is further provided a method for inhibiting the cytokine or biological activity of MIF hi a subject comprising the step of implanting an implantable device according to the invention in the subject. In a yet further aspect, the present invention provides a method of treating, preventing or diagnosing a disease or condition wherein MIF cytokine activity is implicated comprising the step of implanting an implantable device according to the invention in a subject in need thereof.
The present invention further provides an angioplaslic stent for inhibiting the onsel of restenosis, which comprises an angioplasty stent operably coated with a prophylactically effective dose of a composition comprising at least oπe.compound of formula (I).
The present invention further provides a method for inhibiting the onset of restenosis in a subject undergoing angioplasty, which comprises topically administering a stent according to the present invention to the subject at around the time of the angioplasty.
There is further provided a method of reducing the severiLy of stent restenosis in the vicinity of a stent comprising the use of a stent according to the present invention.
BRIEl*1 DESCRIFfION OF THE FIGURES
Figure I shows that treatment with a compound according to the present invention induces a dose-dcpendeπt inhibition of LPS-induced IL-6 production in a mouse macrophage cell line.
Figure 2 shows that treatment with a compound according to the present invention induces a dose-dependent inhibition of IL-I induced COX-2 expression when S112 cells are treated with up to 100 μlvl concentration of compound.
Figure 3A shows that treatment of mice with compound 15 according to the present invention results in a significant dose-dependent suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock.
Figure 3B shows that treatment of mice with compounds 2 and 13 according to die present invention results in a significant dosc-dependenl suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock. Figure 30 shows that treatment of mice with compound 4 according io the present invention results in a significant dose-dependent suppression of LPS-induced serum TNF levels in a mouse model of endotoxic shock.
Figure 3D shows that treatment of mice with compound 19 according to the present invention results hi a significant dose-depeπdcnt suppression of LPS-induccd scrum TNF levels in a mouse model of endotoxic shock.
Figure 4 shows reduction in DTH reactions in vivo in mice treated with compound 13.
Figure 5 shows effect of compound 13 on rhMIF-induced leukocyte adhesion,
DETAILED DKSCmFfION OF THE PREFERKED EMBODIMENTS
In a first aspect, the present invention provides a method of treating, diagnosing or preventing autoimmune diseases, tumours, or chronic or acute inflammatory diseases comprising administering a treatment, prevention or diagnostic effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof wherein:
Figure imgf000018_0001
X is selected from - -O-, -S-, -C(R5)(R5 )- and -N(R0)-;
Y is selected from - -N(R7)-, -O7 -S-, and -C(Ry)2-; Z is selected from >C=O, >C=S, >C-NRG, >S=O and >S(O)2;
R1 is selected from hydrogen, Ci-C3alkyl, (CR5R5OnOR7, C(RsRsOnSR7, (CR5R5OnN(Ro)2 and (CRsRsOnhalo;
R3 is selected from hydrogen, Ci-Qalkyl, (CR16R1 nOnNR11IR15, (CRI6RJ0OPORI7, (CR1ftR1βOpSRπ, (CR,ti6)phalo, (CR1oR1dOpNOj, (CR16R10OnC(O)R2R, (CR1nR16O11S(O)R17, (CR1nR16OnS(O)2R17, (CRK1R16O1IS(O)3R17, and
Figure imgf000019_0001
R4 is selected from hydrogen, halogen, Ci-Cjalkyl, C2-Csalkenyl, C2-C3alkynyl and
Figure imgf000019_0002
each R5 and Ry is independently selected from hydrogen, Ci-Cjalkyl, halov OR7, SR7 and N(Re)2;
each R6 is independently selected from hydrogen, Ci-Cjalkyl and ORy;
each R7 is independently selected from hydrogen and Ci-Cgalkyl;
each R12 and R12* is independently selected from hydrogen, C|-C6alkyl, C2-C6alkenyl, C2- Qalkynyl, OR24, SR24, halo, N(R2^)2, CO2R24, CK NO2, aryl and hcterocyclyl;
each R14 -UId R i3 is independently selected from hydrogen, Ci-C3alkyl, ORn, SRi7, and N(R17^;
each R1ή and R1n' is independently selected from hydrogen, C]-C3alkyl, halo, OR17, SR17 and N(Rn)2;
each Rn is independently selected from hydrogen and Ci-Cjalkyl;
each R1g is independently selected from hydrogen and halo;
R22 is selected from d-Qalkyl, NH2, NH(C,-C6alkyl), N(C,-C6al.kyl)2, OR2y or SR29; each R24 is selected from H and Ci-Cgalkyl;
R2χ is selected from hydrogen, Ci-C(jaLkyl, OR29, SR29 or N(R29)2;
each R2O is independently selected from hydrogen and Ci-Csalkyl;
Q is selected from O, S, NR40, S(O)11 where u is an integer frornl to 2;
R40 is selected from H, OH, and
Figure imgf000020_0001
each R41 and R4T is independently selected from H, OH, halo, NH2, eyanό, and NO2;
R12 is independently selected from H, OR43, COOR4J, CON(R43R4.?), 0(CO)R43, aryl, and heterocyclyl;
each R43 find R43' is independently selected from H, Ci .<j<ilkyl, benzyl, and aryl;
n = O or an integer to 3
m is O or an integer from I to 20;
p is 0 or an integer from 1 to 6;
t is an integer from 1 to 10
v is 0 or an integer from 1 to 10.
In particular, the autoimmune disease, tumour, or chronic or acute inflammatory disease is selected from the group comprising:
rheumatic diseases (including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis) spondyloarthropathies (including but not limited to ankylosing spondylitis, reactive arthritis, Reiter's syndrome), crystal arthropathies (including but not limited to gout, pseudogout, calcium pyrophosphate deposition disease), Lyme disease, polymyalgia rheumatica; connective tissue diseases (including but not limited to systemic lupus erythematosus, systemic sclerosis, polymyositis, derruatomyositis, Sjogren's syndrome);
vasculitides (including but not limited to polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome);
inflammatory conditions including consequences of trauma oc ischaemia;
sarcoidosis;
vascular diseases including atherosclerotic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischaemic heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis;
ocular diseases including uveitis, corneal disease, iritis, iridocyclitis, cataracts; autoimmune diseases (including but not limited to diabetes mellitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis);
pulmonary diseases (including but not limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome);
cancers whether primary or metastatic (including but not limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
renal diseases including glomerulonephritis, interstitial ncpliritis;
disorders of the hypothalamic-pituitary-adrenal axis;
nervous system disorders including multiple sclerosis, Alzheimer's disease; rheumatoid arthritis, cancer), endometrial function (menstruation, implantation, endometriosis);
complications of infective disorders including cndotoxic (septic) shock, cxotoxic (septic) shock, infective (true septic) shock, malarial complications, other complications of infection, pelvic inflammatory disease;
transplant rejection, graft-versus-host disease;
allergic diseases including allergies, atopic diseases, allergic rhinitis;
bone diseases (eg osteoporosis, Paget's disease);
skin diseases including psoriasis, atopic dermatitis, UV(B)-induced dermal cell acLivalion (eg sunburn, skin cancer);
diabetes mcllitus and its complications;
pain, testicular dysfunctions and wound healing;
gastrointestinal diseases including inflammatory bowel disease (including but not limited to ulcerative colitis, Crohn's disease)* peptic ulceration, gastritis, oesophagitis, liver disease (including but not limited to cirrhosis, hepatitis).
MIF cytokine or biological activity is implicated in the above diseases and conditions.
Preferably, flic disease or condition is selected from the group consisting of rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, Crohn's disease, multiple sclerosis, psoriasis, uveitis, diabetes mellitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and chronic obstructive pulmonary disease.
Tn a preferred form Q is S.
In a further preferred form, R40 is C(RnR-H OvRa wherein R.,2 is COOR43. More preferably, R43 is hydrogen or Ci-Cβalkyl, preferably methyl. Li another preferred form, lhe compound of Formula 1 is selected from any one of Compounds 1 to 32 as set out in Lhe Examples herein.
Particularly preferred are Compounds 2, 13 and 19.
As used herein, the term "effective amount" relates to an amount of compound which, when administered according to a desired dosing regimen, provides the desired MIF cytokine inhibiting or treatment or therapeutic activity, or disease/condition prevention. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods. A cytokine or biological activity inhibiting amount is an amount which will at least partially inhibit the cytokine or biological activity of MIF. A therapeutic, or treatment, effective amount is an amount, of the compound which, when administered according to a desired dosing regimen, is sufficient to at least partially attain the desired therapeutic effect, or delay the onset of, or inhibit the progression of or halt or partially or fully reverse the onset or progression of a particular disease condition being treated. A prevention effective amount is an amount of compound which when administered according to the desired dosing regimen is sufficient to at least partially prevent or delay the onset of a particular disease or condition. A diagnostic effective amount of compound is an amount sufficient to bind to !VtIF to enable detection of the MIF-compound complex such that diagnosis of a disease or condition is possible.
Suitable dosages may lie within the range of about 0.1 ng per kg of body weight to 1 g per kg of body weight per dosage. The dosage is preferably in the range of 1 μg to 1 g per kg of body weight .per dosage, such as is in the range of 1 mg to 1 g per kg of body weight per dosage. In one embodiment, the dosage is in the range of 1 mg to 500 mg per kg of body weight per dosage. In another embodiment, the dosage is in the range of 1 mg to 250 mg per kg of body weight per dosage. In yet another preferred embodiment, the dosage is in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per kg of body weight per dosage. In yet another embodiment, the dosage is in the range of lμg to lmg per kg of body weight per dosage. Suitable dosage amounts and dosing regimens can be determined by the attending physician or veterinarian and may depend on the desired level of inhibiting activity, the particular condition being treated, the severity of the condition as well as the general age, health and weighL of lhe subject.
The active ingredient may be administered in a single dose or a series of doses. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a composition, preferably as a pharmaceutical composition.
It will be recognised that other therapeutically active agents such as anti-inflammatory (eg steroids such as glucocorticoids) or anti-cancer agents may be used in conjunction with a compound of Formula (I). Compounds of Formula (1) when administered in conjunction with other therapeutically active agents may exhibit an additive or synergistic effect. These may be administered simultaneously, either as a combined form (ie as a single composition containing the active agents) or as discrete dosages. Alternatively, the other therapeutically active agents may be administered sequentially or separately with the compounds of the invention. Thus, the invention also relates to kits and combinations, comprising a compound of Formula (I) and one or more other therapeutically active ingredients for use in the treatment of diseases or conditions described herein. Without being limiting, examples of agents which could be used in combination with a compound of Formula (I) include: antirheumatic drugs (including but not limited to methotrexate, leflunomide, sulphasalaziπe, hydroxycholorquine, gold salts); immunosuppressive drugs (including but not limited to cyclosporin, mycophenyllate mofetϋ, azathioprine, cyclophosphamide); anti-cytokine therapies (including but not limited to antagonists of, antibodies to, binding proteins for, or soluble receptors for tumor necrosis factor, interleukin 1, interleukin 3, intcrlcukin 5, interleukin 6\ interleukin 8, interleukin 12, interleukin 18, interleukin 17, and other pro-inflammatory cytokines as may be found relevant to pathological states); antagonists or inhibitors of mitogen-activated protein (MAP) kinases (including but not limited to antagonists or inhibitors of extracellular signal-regulated kinases (ERK), the c-Jun N-terminal kinases/slress-aclivaled protein kinases (JNK/SAPK), and thep38 MAP kinases, and other kinases or enzymes or proteins involved in MAP kinase-dependent cell activation); antagonists or inhibitors of the nuclear factor kappa-B (NF-κB) signal transduction pathway (including but not limited to antagonists or inhibitors of I-κB-lcinase, iπterleukin receptor activated kinase, and other kinases or enzymes or proteins involved in NF-κB-dependeπt cell activation); antibodies, protein therapeutics, or small molecule therapeutics interacting with adhesion molecules and co-stimulatory molecules (including but not limited to therapeutic agents directed against intercellular adhesion molecule- 1, CD40T CD40-ligand, CD28, CD4, CD-3, selcctins such as P-setectm or E-selectin); bronehodilators such, as β-adrciioceptor agonists or anticholinergics; antagonists of eicosanoid synthesis pathways such as non-steroidal anti-inflammatory drugs, cyclooxygenase-2 inhibitors, thromboxane inhibitors, orlipoxygena.se inhibitors; antibodies or other agents directed against leukocyte surface antigens (including but not limited to antibodies or other agents directed against CD3, CD4, CD5, CD19, C.D20, I ILA molecules, BLyS); agents used for the treatment of inflammatory bowel disease (including but not limited to sulphasalazine, mesalaziπe, salicylic acid derivatives); anti-cancer drugs (including but not limited to cytotoxic drugs, cytolytic drugs, monoclonal antibodies).
Accordingly, preferably, the compound of formula (I) is administered in conjunction with a second therapeutic agent. More preferably, the second therapeutic agent is a glucocorticoid.
Preferably, the compound of Formula (I) is a compound of Formula (TI) wherein:
Figure imgf000025_0001
π X is selected from - -O-, -S-, -C(R5)(RsO- and -N(R6)-;
Y is selected from - -N(R7)-, -O-, and -S-; Z is selected from >C=O, >C=S, and >C=NR6;
R1 is selected from hydrogen, Ci-G*alkyl, (CR5R5^OR7, C(RsRsOnSR?, CCR5Ry)nN(Rs)2 and (CRsRsOnhalo;
R3 is selected from hydrogen, C1-C6QIlCyI, (CR]6R16OpNR14R1S, (CR1ftR1ή-)POR,7, (CR16R1 ft.)pSR17, (CR16R1ff)phalo, (CR1nR1nOnNO2, (CR16R16^C(O)R28,
Figure imgf000026_0001
(CR16R16OnS(O)2Ri7, (CRi6RKy)nS(O)3R17, and (CRιβRιff)pC(R18)j;
R4 is selected from hydrogen, halogen, Cχ-C3alkyl, C2-C3alk.en.yl, C2.-C3alky-.yl and (CR12RJr)n(CR1 B)3;
each R≤ and Rs> is independently selected from hydrogen, Ci-C/jalkyl, halo, OR7, SR7 and N(Rβ)2;
each Rs is independently selected from hydrogen, Ci-Csalkyl and OR7;
each R7 is independently selected from hydrogen and Ci-Cjalkyt;
each R12 and R^- is independently selected from hydrogen, Ci-Cβalkyl, C2-Coalkenyl, C2- Cβalkynyl, OR2/), SR24, halo, N(R^)2, CO2R24, CN, NO2, aryl and heterocyclyl;
each R14 and R15 is independently selected from hydrogen, CrC3alkyl, OR)7, SRn, and
each R1g and RK,- is independently selected from hydrogen, Cι-C3_ιlkyl, halo, OR17, SRn and N(R1V)2;
each R17 is independently selected from hydrogen and Cι-C3alkyl;
each R1g is independently selected from hydrogen and halo;
R22 is selected from Q-Qalkyl, NH2, NH(Cj-C6al]cyl), N(Ci-C6alkyl)2, OR2y or SR2i,; each R24 is selected from H and Ci-Cr,alkyl;
R2K is selected from hydrogen, Cj-Cealkyl, OR25,, SR29 «r N(R^h;
each R29 is independently se lected from hydrogen and C 1 -C^alkyl;
Q is selected from O , S , S(O)11 where u is an integer from 1 to 2;
R40 is selected from H, OH, and C(R<πR4j')vR42;
each R41 and E4i- is independently selected from H. OH, halo, NH2, CN and NO2;
R^2 is selected from H, OR4:), COOR43, CON(R43R43.), O(CO)R43τ N(R4^R43'), aryl, and heterocyelyl;
each R/i;ι and R43- is independently selected from H, Ci-ealkyl, and benzyl;
n is O or l to 3;
m is O or an integer from 1 to 8;
p is O or an. integer from 1 to 6;
t is an integer from 1 to ,10;
v is 0 or an integer from 1 to 10.
Preferably, the compound of Formula (I) is a compound of Formula (III) wherein:
Figure imgf000027_0001
X is selected from - -O-, -S-, -C(R5)(R5')- and -N(Rfi)-;
Y is selected from - -N(R7)-, -O-, and -S-;
Z is selected from >C=O, >C=S, and >C=NRή;
R1 is selected from hydrogen, C,-C3alkyl, (CR5RSOEOR7, C(R5R5^SR7, (CRSR5OJN(R(V)J and (CR5R5OnIIaIo;
R3 is selected from hydrogen, d-Cealkyl, (CRI6RI6OPNR14RIS, (CR10R1ήOPOR17, (CR1nR16OpSR17, (CR1 0R1 ftOpiialo, (CR1nR16OpNO2, (CR10R16OnC(O)R28, (CR,GR1ft.)nC(=NR24)R22, (CR1nR16OS(O)R17, (CR16R10O11S(O)2R17, (CR16R16OnS(O)3Rn, and (CR16R16θpC(R18)3;
R4 is selected from hydrogen, halogen, d-Cjalkyl, Cz-C^alkenyl, C2-Cjalkyriyl and (CRuR12On(CR1S)3;
each Rj and Ry is independently selected from hydrogen, Ci-C3alkyl, halo, OR7, SR7 and N(R^)2;
each Rf1 is independently selected from hydrogen, Ci-C^alkyl and OR7;
each R7 is independently selected from hydrogen and CrQ^aLkyl;
each R12 and R12' is independently selected from hydrogen, Cι-C6-alkyl, C2-Cή-dkcnyl, C2- Cealkynyl, OR24, SR24, halo, N(R24)^, CO2R2/!, CN, NO2, aryl and heterocyclyl;
each Ru and R15 are independently selected from hydrogen, Ci-C^alkyl, OR17, SR17, and N(Rn)2;
each R]0 and R1^ is independently selected from hydrogen, Ci-C-jalkyl, halo, OR17, SR17 and N(R17J2; each Rn is independently selected from hydrogen and C)-C3alkyl;
each R1s is independently selected from hydrogen and halo;
R22 is selected from C,-C6alkyl, NH2, NH(Q-C6alkyl)τ N(CrC6alkyl)2, OR29 Or SR29;
each RΪ4 is selected from H and C|-C<;alkyl;
R28 is selected from hydrogen, CrCβalkyl, OR29, SRM or N(R^)2;
each R29 is independently selected from hydrogen and Ci-C3alkyl;
R44 is selected from OH,
Figure imgf000029_0001
each R45 and R4S' is independently selected from H, OH, halo, NH2, CN, NO2;
each R/jft is selected from COOR4?, CON(R47R47), 0(CO)R47, N(R47R47);
each R17 and R47' is independently selected from H, Cus alkyl, benzyl;
wherein when v is greater than 1 , ~Rά(, can be OR471
wherein when v is greater than 1 , R4g can be 11;
π is U or 1 to 3;
m is O or an integer from 1 to 8;
p is O or an integer from 1 to 6;
t is an integer from 1 to 10;
v is O or an integer from 1 to 10.
tn a second aspect, the present invention provides a compound of Formula (II) or a pharmaceutically acceptable suit or prodrug thereof wherein:
Figure imgf000030_0001
II
X is selected from - -O-, -S-, -C(R5)(R5 1)- md -N(Rn)S
Y is selected from - -N(R7)-, -O-, and -S-;
Z is selected from >C=O, >C=S, and >C=NR6;
R1 is selected from hydrogen, C,-C3alkyl, (CR5R5OHOR7, C(R5R5^SR7, (CR5RsOnN(Rn)2 and (CR5R5-X1IIaIo;
R;} is selected from hydrogen, Ci-C6alky], (CR1nR16OpNR14R1S, (CRK,R160pQR-i7, (CRwR1«0pSR17, (CR16R1ff)phalo, (CR16R16OpNO2, (CR16Rw)1AOJR28,
Figure imgf000030_0002
(CRi6R16-)aS(O)R17, (CR1nR16OnS(O)2R17, (CR10R1nOnS(O)3R17, and (CR16R1COpC(R1S).);
R4 is selected from hydrogen, halogen, C|-C3.ιlkyl, Ca-Cjalkenyl, C:2-C3alkynyl and
Figure imgf000030_0003
each Rs and Ry is independently selected from hydrogen, Ci-C3alkyl, halo, OR7, SR7 and N(Re)2;
each R6 is independently selected from hydrogen, C|-C^alkyi and OR7;
each Rη is independently selected from hydrogen and Q-Csalkyl; each R12 and Rιτ is independently selected from hydrogen, Ci-C^lkyl, C2-C6alkenyl, Ca- Cήalkyny], OR24, SR24, halo, N(R24)., CO2R24. CN, NO2, aryl and heterocydyl; .
each Ru and R15 is independently selected from hydrogen, Cj-Osalkyl, OR17, SRw> and N(R, 7)2;
5 each is independently selected from hydrogen, Ci-Csalkyl, halo, OR)7, SR17 and
each R.J7 is independently selected from hydrogen and Ci-Cjalkyl;
each R1g is independently selected from hydrogen and halo;
R22 is selected from CrC<jalkyt, NH21 NH(Cι-C6alkyI), N(Ci-C<5ali.yl)2, OR2y or SR29;
ϋ each R24 is selected from H and Ci -Cgalkyl;
Ra? is selected from hydrogen, Cj-Coalkyl, OR2c», SR29 or N(R29)2 *,
each R2^ is independently selected from hydrogen and Ci-Caalkyl;
Q is selected from O 1 S , S(O)U where u is an integer from 1 to 2;
R40 in selected from H, OH, and C(R4iR-n-)vR42;
5 each R41 and R41' is independently selected from H, OH, halo, NHo, CN and NO2;
R42 is selected firom H, OR43, COOR43, CON(R4^R430, 0(CO)R4S, N(R4^y), aryl, and heterocyclyl; .
each R43 and R*r is independently selected from H, Cut, alkyl, and benzyl;
n is O or 1 to 3;
0 m is O or an integer from ] to 8; p is ϋ or an integer from 1 to 6;
t is an integer from 1 to 10;
v is 0 or nn integer from 1 to 10
provided that the compound is not
Figure imgf000032_0001
In a third aspect, the present invention provides a compound of Formula III or a pharmaceutically acceptable salt or prodrug thereof wherein:
Figure imgf000032_0002
111
X is selected from - -O-, -S-, -C(Rs)(R5')- and -N(R6)-;
Y is selected from - -N(R7)-, -O-, and -S-; R1 is selected from hydrogen. C|-Q>alkyl, (CR5R^)nOR7, C(R5R5OnSR7, (CR5Ry)nN(Re)2 and (CR3Ry)11-IaIo;
R3 is selected from hydrogen, Cj-Qalkyl, (CR1βR1fiOpNRμR1s, (CR16R I6OpORn. (CR^R1βOμSRn, (CR^R1sOphalo, (CR15R16OpNO2, (CR16ft-)nC(O)R2s, (CR16RIn)1A=NR24)R22, (CR16R16OS(O)R17, (CR1^w)nS(O)2R17. (CR16R1S)nS(O)3R17, and (CR16R16OpC(R1K)3;
R4 is selected from hydrogen, halogen, C,-C3alkyl, Ci-C^alkenyl, Cz-Cjatkyαyl and (CR12RnO11(CR18)Ir,
each R,i and R51 is independently selected from hydrogen, C1-C3HUCyI, halo, OR7, SR7 and N(R6)2;
each Ra is independently selected from hydrogen, Ci-Cjalkyl and OR7;
each R7 is independently selected from hydrogen and Ci-Cjalkyl;
each R12 and R12' is independently selected from hydrogen, Cj-CoaLkyl, Cϊ-Cgalkcnyl, C2- Cftalkynyl, OR2^, SR24, halo, N(R34)2, CO2R14, CN, NO2, aryl aud heterocyclyl;
each RK and R1S are independently selected from hydrogen, Ci-C3a!kyt, OR17, SR17, and N(R17)U
each R1g and R1n- is independently selected from hydrogen, Ci-Qjalkyl, halo, OR17, SR17 and N(RI7)2\
each R17 is independently selected from hydrogen and Ci-C-3alkyl;
each R1 x is independently selected from hydrogen and halo;
R22 is selected from C|-Caalkyl, NH2, NH(C,-C6alkyl), N(Ci-C6alkyl)2, OR2^i or SR29!
each R2/1 is selected from H and d-Qalkyl; R2H is selected from hydrogen, Ci-Coalkyl, OR29, SR2y or N(R29h;
each R.29 is independently selected from, hydrogen and Ci-C3alkyl;
R44 is selected from OH,
Figure imgf000034_0001
each R45 and
Figure imgf000034_0002
is iπdei>endently selected from H, OH, halo, NH2, CN, NO2;
each R4n is selected from COOR47, CON(R47R47'), 0(CO)R47, N(R47R4?');
each R47 and R47' is independently selected from H, Ci-6 af kyl, benzyl;
wherein when v is greater than 1 , R46 can be OR47;
wherein when v is greater than 1 , R4^ can be H;
n is O or 1 to 3;
m is O or an integer from 1 to 8;
p is O or an integer from Ho 6;
t is an integer from. 1 to 10;
v is 0 or an integer from 1 to 10
provided that the compound is not
Figure imgf000034_0003
As used herein, the term "alkyl" refers to monovalent straight, branched or, where appropriate, cyclic aliphatic radicals, having 1 to 3, I to 6, 1 to 10 or 1 to 20 carbon atoms, e.g. methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, π-butyl, sec-butyl, t-butyl and cyclobutyl, n-pentyl, 1-methylbutyl, 2-methylbυtyl, 3-methylbutyl, cyclopcntyl, n-hcxyl, 1- 2- 3- or 4- methylpcntyl, 1- 2- or 3-cthylbulyl, 1 or 2- propylpropyl or cyclohexyl.
An alkyl group may be optionally substituted one or more times by halo (eg chloro, lluoro or bromo), CN, NO2, CO2H, CQA-βalkyl, CO2NH2, CO2NH(C !-fialkyl), CO2N(CI-6aIkyl)2, OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl , benzyloxy, phenoxy, NH2,
NH(Ci.6alkyl) or N
Figure imgf000035_0001
A preferred optional substituent is a polar substituent. Examples of alkoxy include methoxy, ethoxy, n-propoxy, iso-propoxy, cyclopropoxy, and butoxy (n-, sec- 1- and cyclo) pentoxy and hexyloxy. The "alkyl" portion of an alkoxy group may be substituted as described above,
As used herein, the term "alkenyl" refers to straight, branched, or where appropriate, cyclic carbon containing radicals having one or more double bonds between carbon atoms. Examples of such radicals include vinyl, allyl, butcnyl, or longer carbon chains such as those derived from palmitoleie, oleic, linoleic, linolenic or arachidonic acids. An alkenyl group may be optionally substituted one or more tunes by halυ (eg chloro, f luoro or bromo), CN, NO2, CO2H,
Figure imgf000035_0002
CO2NH2, CO3NH(C1 6alky0, CO2N(C J-salkyl)2, OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl, benzyloxy, phenoxy, NHi,
Figure imgf000035_0003
or N(Ci-SaUCyI)2. Λ prelarred optional substituent is a polar substituent.
As used herein, the term "alkynyl" refers to straight or branched carbon containing radicals having one or more triple bonds between carbon atoms. Examples of such radicals include propargyl, butynyl and hexynyl. An alkynyl group may be optionally substituted one or more times by halo (eg chloro, fluoro or bromo), CN, NO2, CO2H,
Figure imgf000035_0004
COaNH2, CQ2NH(C1.6alkyl),
Figure imgf000035_0005
OH, alkoxy, acyl, acetyl, halomethyl, trifluoromethyl, benzyloxy, phenoxy. NI I2, NH(Ci. oalkyl) or N(Ci_salkyl)2. Λ preferred optional substituent is a polar substituent.
Examples of suitable NI I(alkyl) and N(alkyl>2 include methylamino, ethylamino, isopropylamiπo, dimethylamiπo, n-propylamino, diethylamino and di-isopropylamino.
The term "halogen" (or "halo") refers to fluorine (fluoro), chlorine (chloro), bromine (bromo) An aryl gtoup, as used herein, refers to Cβ-Cio aryl groups such as phenyl or naphthalene. Aryl groups may be optionally substituted one or more times by halo (eg, chloro, fluoro or btoπώ), CN, NO2, CO2H, CO2Ci-6alkyl, CO2NH2, COnNH(C1. oalkyl), CO2N(Ci^aIkJl)2, OH, alkoxy, acyl, acetyl, halomethyl, trifluorornethyl, benzyloxy, phenoxy, NH2, NH(C1 oalkyl) or N(Ci.6alkyl)2.
As used herein, the term, "hctcrocyclyl" refers to a cyclic, aliphatic or aromatic radical containing at least one heteroatom independently selected from O, N or S, Examples of suitable heterocyclyl groups include fury], dioxolanyl, dioxanyl, dithiaiiyl, dithiolanyl, pyridinyl, pyrimidinyl, pyrazolyl, piperidiiiyl, pyrrolyl, thyaphenyl, oxazotyl, imidazolyl, thjazolyl, ϊsoxazolyl, isothiazolyl, quinolyl, isoquinolyl, indolyl, betLtϋfuranyl, benzolhiophenyl, triazolyl, tetrazolyl, oxadiazolyl and puriπyl. Heterocyclyl groups may be optionally substituted one or more times by halo (eg, cliloro, fluoro or bromo), CN, NO2, CO2H, CO2C^alkyl, CO2NII2, CO2NH(Ci.6alkyl), CO2N(C,.ήalkyl)2j OH, alkoxy, acyl, acetyl, halomethyl, trifluoromelhyl, benzyloxy, phenoxy, NH2, NH(Ci -salkyl) or N(C1-fiaIkyl)2.
The term "salt, or prodrug" includes any pharmaceutically acceptable salt, ester, solvate, hydrate or any oilier compound which, upon administration to the recipient is capable of providing (directly or indirectly) a compound of Formula (1) as described herein. The term "pro-drug" is used in its broadest sense and encompasses those derivatives that, arc converted in vivo to the compounds of the invention. Such derivatives would τeadily occur to those skilled in the art, and include, for example, compounds where a free hydroxy group is converted into an ester, such as an acetate, or where a free amino group is converted into an amide. Procedures for acylating hydroxy or amino groups of the compounds of the invention are well known in the art and may include treatment of the compound with an appropriate carboxylic acid, anhydride or acylchloridc in the presence of a suitable catalyst or base.
Suitable pharmaceutical Iy acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids such as hydrochloric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic, and hydrobromic acids, or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, maleic, citric, lactic, muck, gluconic, benzoic, succinic, oxalic, phenyl acetic, methanesulphonic, tolucnesulphonic, benezenesulphonie, salicyclic sulphanilic, aspartic, glutamic, edetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.
Base salts include, but are not limiled to, those formed with pharmaceutically acceptable cations, such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylanimonium.
Basic nitrogen-containing groups may be quarternised with such agents as lower alkyl halide, such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl and diethyl sulfate; and others.
It will also be recognised that some compounds of formula (I) may possess asymmetric centres and arc therefore capable of existing in more than one stereoisomer^ form. The invention thus also relates to compounds in substantially pure isomeric form at one or more asymmetric centres eg., greater than about 90% cc, such as about 95% or 97% ee or greater than 99% ce, as well as mixtures, including racemic mixtures, thereof. Such isomers may be prepared by asymmetric synthesis, for example using chiral intermediates, or by chiral resolution.
Λ further aspect of the invention provides for the use of a compound of Formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment of a disease or condition as above.
In a further aspect of the invention, there is provided a pharmaceutical composition comprising a compound of formula. (I) together with a pharmaceutically acceptable carrier, diluent or excipieπt.
The formulation of such compositions is well known to those skilled in the art. The composition may contain pharmaceutically acceptable additives such as carriers, diluents or excipicnts. These include, where appropriate, all conventional solvents, dispersion agents, fillers, solid earners, coating agents, antifungal and antibacterial agents, dermal penelralion agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
The carrier must be pharmaceutical Iy acceptable in the sense of being compatible with the other ingredients of the composition and not injurious to the subject. Compositions include those suitable for oral, rectal, Lnhalational, nasal, transdermal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intraspinal, intravenous and intradermal) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
Depending on the disease or condition to be treated, it may or may not be desirable for a compound of Formula (I) to cross the blood/brain barrier. Thus the compositions for use in the present invention may be formulated to be water or lipid soluble.
Compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, sachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in- water liquid emulsion or a watcr-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder ov granules, optionally mixed with a binder (eg inert diluent, preservative, disinlegraπt (eg. sodium starch glyc-olate, cross-linked polyvinyl ovrrolidone, cross-linked sodium carboxvrπelhvl cellulose1)1) surface- active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methyl cellulose in varying proportions to provide the desired release profile. Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
Compositions suitable for topical administration in the mouth iaclude lozenges comprising the active ingredient in a flavoured base, usually sucrose and acacia or tragacanth gum; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia gum; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
The compounds of Formula (1) may also be administered intranasal Iy or via inhalation, for example by atomiser, aerosol OΪ nebulizer means.
Compositions suitable for topical administration to the skin may comprise the compounds dissolved or suspended in any suitable carrier or base and may be in the form of lotions, gel, creams, pastes, ointments and the like. Suitable carriers include mineral oil, propylene glycol, polyoxycthylene, polyoxypropylene, emulsifying wax, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodeeanol, benzyl alcohol and water. Transdermal devices, such as patches, may also be used to administer the compounds of the invention.
Compositions for rectal administration may be presented as a suppository with a suitable carrier base comprising, for example, cocoa butter, gelatin, glycerin or polyethylene glycol.
Compositions suitable for vaginal admin istration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate. Compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain anti-oxidants, buffers, bactericides and solutes which, render the composition isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from slerilc powders, granules and tablets of lhe kind previously described.
Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the active ingredient.
It should be understood that in addition to the active ingredients particularly mentioned above, the compositions of this invention may include other agents conventional in the art having regard to the type of composition in question, for example, those suitable for oral administration may include such further agents as binders, sweeteners, thickeners, flavouring agents, disintegrating agents, coating agents, preservatives, lubricants and/or time delay agents. Suitable sweeteners include sucrose, lactose, glucose, aspartame or saccharine. Suitable disintegrating agents include corn starch, methylcellulose, polyvinylpyrrolidone, xaήthan gum, bentonitc, alginic acid or agar. Suitable flavouring agents include peppermint oil, oil of wiiilergreen, cherry, orange or raspberry flavouring. Suitable coating agents include polymers or copolymers of acrylic acid and/or methacrylic acid and/or their esters, waxes, fatty alcohols, zcifl, shellac or gluten. Suitable preservatives include sodium bcnzoate, vitamin E, alpha-tocopherol, ascorbic acid, methyl paraben, propyl paraben or sodium bisulphite. Suitable lubricants include magnesium stearate, stearic acid, sodium oleatc, sodium chloride or talc. Suitable time delay agents include glyceryl monostearate or glyceryl distearate.
In a further aspect, the present invention provides a method of inhibiting cytokine or biological activity of MIF comprising contacting MIK with a cytokine or biological activity inhibiting effective amount of a compound of formula (I), or a pharmaceutically acceptable salt Of prodrug thereof.
In another aspect, the invention provides a method of treating, preventing or diagnosing a disease or condition wherein MlF cytokine or biological activity is implicated comprising the administration of a treatment, prevention or diagnostic effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof.
In a further aspect, there is provided the use of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment, prevention or diagnosis of a disease or condition wherein MlF cytokine or biological activity is implicated.
As used herein, MIF includes liuinaα or other animal MlF and derivatives and naturally occurring variants thereof which at least partially retain MIF cytokine or biological activity. Thus, the subject to be treated may be human or other animal such as a mammal. Non-human, subjects include, but are not limited to primates, livestock animals (eg sheep, cows, horses, pigs, goats), domestic animals (eg dogs, cats), birds and laboratory test animals (eg mice. rats, guinea pigs, rabbits). JMHF is also expressed in plants (thus "MIF" may also refer to plant MIF) and where appropriate, compounds of Formula (I) may be used in botanical/agricultural applications such as crop control.
Reference herein to "cytokine or biological activity" of MIF includes Lhe cytokine or biological effect on cellular function via autocrine, endocrine, paracrine, cytokine, hormone or growth factor activity or via intracellular effects.
In another aspect, -he invention provides a method of treating or preventing a disease or condition wherein MIF cytokine or biological activity is implicated comprising:
administering to a mammal a compound of formula (I) and a second therapeutic agent. In a preferred embodiment of this aspect of the invention, the second therapeutic agent Is a glucocorticoid compound.
In another aspect, the present invention provides a method of prophylaxis or treatment of a disease or condition for which treatment with a glucocorticoid is indicated, said method comprising: administering to a mammal a glucocorticoid and a compound of formula (1).
In yet another aspect, the present invention provides a method of treating steroid-resistant diseases comprising administering to a mammal a glucocorticoid and a compound of formula (I).
In a further aspect, the present invention provides a method of enhancing the effect of a glucocorticoid in mammals comprising administering a compound of formula (I) simultaneously, separately ov sequentially with said glucocorticoid.
In yet a further aspect, the present invention provides a composition comprising a glucocorticoid and a compound of formula (I).
In a further aspect of the invention there is provided a use of a glucocorticoid in the manufacture of a medicament for administration with a compound of formula (I) for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
In yet a further aspect of the invention there is provided a use of a compound of formula (I) in the manufacture of a medicament for administration with a glucocorticoid for the treatment or prophylaxis of a disease or condition for which, treatment of a glucocorticoid in indicated.
In yet a further aspect of the invention there is provided a use of a glucocorticoid and a compound of formula (I) in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
Preferably the amount of glucocorticoid used in the methods, uses and compositions of the invention is less than the amount which would he effective ϊa the absence of the compound of formula (I). In the treatment of steroid-resistant diseases or conditions- which are not responsive, to glucocorticoids, any amount of glucocorticoid which is effective in combination with a compound of formula (I) is considered less than the amount which would be effective in the absence of a compound formula (I). Accordingly, the invention provides a steroid-sparing therapy.
The term "disease or condition for which treatment with a glucocorticoid is indicated" refers to diseases or conditions which are capable of being treated by admmistraiion of a glucocorticoid including but not limited to autoimmune diseases, tumours, or chronic or acute inflammatory diseases. Examples of such diseases or conditions include:
rheumatic diseases (including but not limited to rheumatoid arthritis, osteoarthritis, psoriatic arthritis) spondyloarthropathies (including but not limited to ankylosing spondylitis, reacLive arthritis, Reiter's syndrome), crystal arthropathies (including but not limited to gout, pscudogoul, calcium pyrophosphate deposition disease), Lyme disease, polymyalgia rheumatica;
. connective tissue diseases (including but not limited to systemic lupus erythematosus, systemic sclerosis, polymyositis, dermatomyositis, Sjogren's syndrome);
vasculilides (including but not liirήLedto polyarteritis nodosa, Wegener's granulomatosis, Churg-Strauss syndrome);
inflammatory conditions including consequences of trauma or ischaetnia;
sarcoidosis;
vascular diseases including atherosclerotic vascular disease and infarction, atherosclerosis, and vascular occlusive disease (including but not limited to atherosclerosis, ischacniie heart disease, myocardial infarction, stroke, peripheral vascular disease), and vascular stent restenosis; 43.
autoimmune diseases (including but not limited to diabetes mellitus, thyroiditis, myasthenia gravis, sclerosing cholangitis, primary biliary cirrhosis);
pulmonary diseases (including but not limited to diffuse interstitial lung diseases, pneumoconioses, fibrosing alveolitis, asthma, bronchitis, bronchiectasis, chronic obstructive pulmonary disease, adult respiratory distress syndrome);
cancers whether primary or metastatic (including but not limited to prostate cancer, colon cancer, lymphoma, lung cancer, melanoma, multiple myeloma, breast cancer, stomach cancer, leukaemia, cervical cancer and metastatic cancer);
renal diseases including glomerulonephritis, interstitial nephritis;
disorders of the hypothalamic-pituitary-adrenal axis;
nervous system disorders including multiple sclerosis, Alzheimer's disease;
diseases characterised by modified angiogcncsis (eg diabetic retinopathy, rheumatoid arthritis, cancer), endometrial function (menstruation, implantation, endometriosis);
complications of infective disorders including endotoxic (septic) shock, exotoxic
(septic) shock, infective (true septic) shock, malarial complications, other complications of infection, pelvic inflammatory disease;
transplant rejection, graft- vcrsus-host disease;
allergic diseases including allergies, atopic diseases, aHeTgic rhinitis;
bone diseases (eg osteoporosis, Pagct's disease);
skin diseases including psoriasis, atopic dermatitis, UV(B)-induccd dermal cell activation (eg sunburn, skin cancer); pain, testicular dysfunctions and wound healing;
gastrointestinal diseases including inllammaiory bowel disease (including but not limited to ulcerative colitis, Crohn's disease), peptic ulceration, gastritis, oesophagitis, liver disease (including but nol limited to ciπhosis, hepatitis).
These diseases or conditions may also include steroid-resistant diseases or conditions where treatment with a glucocorticoid is indicated, bat where the glucocorticoid is ineffective or is not as effective as expected.
The methods of the invention are preferably performed in a steroid-sparing manner. The term "steroid-sparing" refers to a combination therapy method that allows a reduction in the amount of glucocorticoid administered while still providing an effective therapy for the disease or condition being treated or prevented.
Steroid-resistant diseases or conditions are diseases or conditions for which treatment with a glucocorticoid is indicated, but where the glucocorticoid is ineffective or is not as effective as expected. This term encompasses diseases or conditions for which the effective dose of glucocorticoid results in unacceptable side effects and/or toxicity. Some steroid-resistant diseases or conditions may require a dosage of glucocorticoid so large that they are considered non-responsive and therefore arc not ahlc to be successfully treated with glucocorticoids. Some steroid-resistant diseases or conditions may require a large dosage of glucocorticoid to achieve only a small effect on the symptoms of the disease or condition. Furthermore, some patients, diseases or conditions present with symptoms that do not respond to treatment with a glucocorticoid, or may become less sensitive to glucocorticoid treatment over time.
Glucocorticoids are a group of steroid hormones, which are used to treat or prevent a wide range of diseases or conditions. Suitable glucocorticoids may be synthetic or naturally occurring and include but are not limited to prednisolone, prednisone, cortisone acetate, beclamethasone, fluticasone, hydrocortisone, dexamethasone, methyl prednisolone, triamcinolone, budesoπide and betamethasone. In preferred embodiments of the invention, the glucocorticoid used is selected from prednisone, prednisolone, hydrocortisone, fluticasone, betamethasone, betamethasone, methyl prednisolone, budesonide, triamcinolone, dexainelhasone and cortisone. Most preferably, the glucocorticoid is selected from prednisone, prednisolone, methyl prednisolone, fluticasone and beclaincthasone, Beclameth&sone and fluticasone are particularly preferred for treating asthma. Prednisone, prednisolone and methyl prednisolone are particularly preferred in the treatment of systemic or local inflammatory diseases.
The amounts of glucocorticoid and compound of formula (I) are selected such that in combination they provide complete or partial treatment or prophylaxis of a disease or condition for which a glucocorticoid is indicated. The amount of compound formula (I) Ls preferably an amount that will at least partially inhibit the cytokine or biological activity of MIF. The amount of glucocorticoid is preferably less than the amount required in the absence of the compound of formula (I). The amounts of glucocorticoid and compound of formula (I) used in a treatment or therapy are selected such that in combination they at least partially attain the desired therapeutic effect, or delay onset of, or inhibit the progression of, or halt or partially or fully reverse the onset or progression of the disease or condition being treated. The amounts of glucocorticoid and compound of formula (I) used in the prophylaxis of a disease or condition arc selected such that in combination they at least partially prevent or delay the onset of the .disease or condition. Dosing may occur at intervals of minutes, hours, days, weeks, months or years or continuously over any one of these periods.
Suitable doses of a compound of formula (I) may lie within the range of about 0.1 πg per kg of body weight to 1 g per kg of body weighr per dosage. The dosage is preferably in the range of 1 μg to 1 g per kg of body weight per dosage, such as is in the range of 1 mg to 1 g per kg of body weight per dosage. In one embodiment, the dosage is in the range of 1 mg to 500 mg per kg of body weight per dosage. In another embodiment, the dosage is in the range of 1 mg to 250 mg per kg of body weight per dosage. In yet another preferred embodiment, the dosage is in the range of 1 mg to 100 mg per kg of body weight per dosage, such as up to 50 mg per kg of body weight per dosage, In yet another embodiment, the dosage is in the range of lμg to lrng per kg of body weight per dosage.
Suitable dosage amounts of glucocorticoids will depend, in part, on lhe mode of administration and whether the dosage is being administered in a single, daily or divided dose, or as a continuous infusion. When administered orally, intravenously, intramuscularly, intralesionally or intracavity (eg. intra-articular, intrathecal, intrathoracic), dosages are typically between 1 mg to lOOO nig, preferably 1 mg to 100 mg, more preferably 1 mg to 50 mg or 1 mg to 10 mg per dose. When administered topically or by inhalation as a single, daily or divided dose, dosages are typically 1 ng to 1 μg, 1 ng to 1 mg oτ 1 pg to 1 μg.
Suitable dosage amounts and dosing regimens cau be determined by the attending physician or veterinarian and may depend on the desired level of inhibiting activity, the particular condition being treated, the severity of the condition as well as the general age, healLh and weight of the subject.
The glucocorticoid and compound of formula (I) 'may be administered simultaneously or sequentially. The active ingredients may be administered alone but are preferably administered as a pharmaceutically acceptable composition or separate pharmaceutically acceptable compositions.
The formulation of such compositions is well known to those skilled in lhe art and are described above in relation to compounds of formula (I). The composition or compositions may contain pharmaceutically acceptable additives such as carriers, diluents or excipients. These include, where appropriate, all conventional solvents, dispersion agents, fillers, solid carriers, coating agents, antifungal and antibacterial agents, dermal penetration agents, surfactants, isotonic and absorption agents and the like. It will be understood that the compositions of the invention may also include other supplementary physiologically active agents.
Preferred unit dosage compositions are those containing a daily dose or unit, daily sub-dose, as herein above described, or an appropriate fraction thereof, of the glucocorticoids and/or The compounds of formula (I), either as the only active agent oc together with another active agent, eg; a glucocorticoid, may also be presented for use in veterinary compositions. These may be prepared by any suitable means known in the' ait. Examples of such compositions include those adapted for:
oral administration, external application (eg drenches including aqueous and non-aqueous solutions or suspensions), tablets, boluses, powders, granules, pellets for admixture with feedstuiϊs, pastes for application to the tongue;
parenteral administration, eg subcutaneous, intramuscular or intravenous injection as a sterile solution or suspension; and
topical application eg creams, ointments, gels. lotioiLS, etc.
By virtue of their ability to bind to or antagonize MIF, compounds of Formula (I) or salts or derivatives thereof may be used us laboratory or diagnostic or in vivo imaging reagents. Typically, for such use the compounds would be labelled in some way, for example, radio isotope, fluorescence or colorknetric labelling, or be chelator conjugated. In particular, compounds of Formula (!) could be used as part Of an assay system for MIF or as controls in screens for identifying other inhibitors. Those skilled in the art are familiar with such screens and could readily establish such screens using compounds of Formula (I). Those skilled in the art will also be familiar with the use of chelate conjugated molecules for in vivo diagnostic imaging.
Inhibitors of MlF may also b& used in implantable devices such as stents. Accordingly, in a further aspect the present invention provides an implantable device, preferably a stent, comprising:
(i) a reservoir containing at least one compound of formula (I); and
(ii) means to release or elute the inhibitor from the reservoir There is further provided a method Tor inhibiting the cytokine or biological activity of MIF in a subject comprising the step of implanting an implantable device according to the invention in the subject.
Preferably, the method is for inhibiting the cytokine or biological activity of MIF in a local region of the subject and the device is implanted within or proximate to die local region of the subject.
In a yet further aspect, the present invention provides a method of treating, preventing or diagnosing a disease or condition wherein MIF cytokine activity is implicated comprising the step of implanting an implantable device according to the invention in a subject in need thereof.
Preferably, the disease or condition is confined to a local region of the subject and the device is implanted with in or proximate to the local region.
The present invention further provides an angioplasty stent, for inhibiting the onset of restenosis, which comprises an aπgioplastic stent Qperably coated with a prophylactically effeclive dose of a composition comprising at least one compound of formula (T).
Aπgioplastic stents, also known by other terms such as "intravascular stents" or .simply "stents", are well known in the art. They arc routinely used to prevent vascular closure due to physical anomalies such as unwanted inward growth of vascular tissue due to surgical trauma. They often have a tubular, expanding lattice- type structure appropriate for their function, and can optionally be biodegradable.
In this invention, the stent can be operably coated with at least one compound or formula (I) using any suitable means known in the art. Here, "operably coating" a stent means coating it in a way that permits the timely release of the compottαd(s) of formula ( I) into the surrounding tissue to be treated once the coated stent is administered. Such coating methods. for example, can use the polymer polypyrrole. The present invention farther provides a method for inhibiting the onset of restenosis in a subject undergoing angioplasty, which comprises topically administering a stent according to the present invention to the subject at around the time of the angioplasty.
As used herein, administration "at around the time of angioplasty" can be performed during the procedure, or immediately before or after the procedure. The administering can be performed according to known methods such as catheter delivery.
There is further provided a. method of reducing the severiLy of stent restenosis in the vicinity of a stent comprising the use of a stent according to flic present invention.
The construction of stents that, release or elutc a pharmaceutical active is known to those skilled in the art. The standard approach is to use current highly refined metallic stent designs with polymer materials that release the active in a controlled manner. Several polymer materials have been used for the coaling of stents to permit the eiution of drugs. These include bioerodible polymers such as poly-L lactic acid, biostable polymers such as polyurcthanc derivatives and slilicone-based polymers, as well as hydrogels. It will be recognised by those skilled in the art that the function of a drug-eluting stent requires the drug io bo bound to ihe stent or its polymer or other coating in such a way as to allow steady release of drug over a period of time, and that the drug is able to be locally absorbed into cells in the vessel and stent lumen. The optimum stent coating material and delivery parameters vary according to the tissue retention of the drug, such that rapid release of a tissue-retained drug can have long lasting effects, whereas a drug retained in tissues for a shorter time would need to be released over a longer period. A person skilled in the art would be able to select, appropriate materials and conformations of stent for a particular purpose and particular small molecule inhibitor.
Proposed Methods of Synthesis
Commercially available starting materials for the preparation of examples include the unsubstitutcd hctcrocyclcs where X and Y are a combination of CHt, O, NH and S (sec Scheme 1). In cases where Z is C=O those include; beπ7.imidazol-2-one (2- (2-bcnzoxai-olinoπe) and betizothiazol-Z-onc (2-hydroxybenzothiazole). In cases where Z is C=NIl these include the tautomeric compounds; 2-ainmoben7,imidazoIe, 2- aπiitiobenzαthiazσle and 2-amiπobenzoxazole. Purther elaboration of the heterocyclic ring may be made by alkylation of basic functionalities using reagents such as methyl iodide or dimethyl sulfate in the presence of base.
Friedel -Crafts acylatioπ of these hctcrocycles with haloalkyl acid halides and aluminium chloride in solvents such as 1,2-dichloroctliatie or Λ^ΛΛdimetl-ylformarαide woidd afford a range of haloalkyl ketones as shown in Scheme 1. The haloalkyl acid halides for t = 1-5 are available commercially, while longer homologues may be prepared by treatment of the more widely available hydroxy-acids with a combination of HBr and oxalyl chloride, or by treatment with thionyl chloride.
Cl(CH2JtCOCI , AICt3
Figure imgf000051_0002
1,2-dichloroethane
Figure imgf000051_0001
Scheme 1
Displacement of the halogen witli an appropriately functionalizcd sulfur or nitrogen nuclcophile in the presence of a noα-nuclcophilic base would give rise to a range of examples where Q is NH or S. iα cases where a nitrogen nuclcophile is used this could be either a primary or secondary amine, affording secondary or tertiary amine examples respectively, In cases where a sulfur πucleophile is used oxidation of the resulting sulfide with reagents such as hydrogen peroxide would generate sulfoxide examples (u =1). Further oxidation using a stronger oxidant such as potassium permanganate or an additional equivalent of hydrogen peroxide would give rise to sulfone examples (u = 2) (see Scheme 2).
Figure imgf000052_0001
Scheme 2
Displacement of the halogen by oxygen nuclcophiles can be achieved by suitable protection, if necessary, of any additional functionality on the alcohol, followed by treatment with sodium hydride or sodium metal to generate the more nudeophilϊc alkoxide anion. This procedure would allow access to the range of examples thai haye Q = O (sec Scheme 3).
Figure imgf000052_0002
Scheme 3
ff in place of the haloalkyl acid halides shown in Scheme 1, cyclic anhydrides or alkoxycarbonyj acid halides are used, then a scries of keto-aάds may be prepared (Scheme 4). Selective reduction of the ketone functionality can be achieved with a selection of reagents including; zinc amalgam, tricthylsilane and sodium borohydride, to afford the corresponding cfu'boxylic acids.
Figure imgf000053_0001
reduction
Figure imgf000053_0002
Scheme 4
Conversion of the acid to the acid chloride- followed by LreatmeiU with cliazomethane and then HBr, affords bromoalkyl ketones (t = 1 ) which can, be used to prepare examples as previously illustrated in Schemes 2 (Q = N, S) and 3 (Q = O),
As described above, compounds of Formula (I) may be prepared using the methods depicted or described herein or known in the art. It. will be understood that minor modifications to methods described herein or known in the art may be required to synthesize particular compounds of Formula (I). General synthetic procedures applicable to the synthesis of compounds may be found in standard references such as Comprehensive Organic
Transformations, R. C. Larock, 1989, VCH Publishers and Advanced Organic Chemistry, J. March, 4th Edition (1992), Wiley InierScience, and references therein. Tt will also be recognised that certain reactive groups may require protection and dcproiection during the synthetic process. Suitable protecting and deprotecting methods for reactive functional groups are known in the art for example in Protective Groups iii Organic Synthesis, T. W. Green & P. Wife, John Wiley & Son, 3rd Edition, 1999. In order that the nature of the present invention may be more clearly understood, preferred forms thereof will now be described with reference to the following non-limiting examples.
SYNTHETIC EXAMPLES
General Experimental
Melting points are uncorrcctcd. Proton nuclear magnetic resonance (' H nmr) spectra were acquired on cither a Bruker Avancc 300 spectrometer at 300 MHz, or on a Varian Inova spectrometer at 400 MHz, using the duetcrated solvents indicated. Low resolution mass spectrometry analyses were performed using a Mieromass Platform II single quadrapole mass spectrometer equipped, with an electrospray (ESl) or atmospheric pressure chemical ionization (APCI) ion source. Sample management was facilitated by an Agilent 1100 series HPLC system.
Commercially sourced starting materials and solvents were used without furtlicr purification.
The following abbreviations have been used: tap, melting point; DCB, 1,2-dichloroethane; DMF, N.N-diiiicthylformamide; THF, tetrahydrofuran; TLC, thin layer chromatography; S-O2, silica gel; dniso, ditnelhylsulfoxide; DCM, dichloromelhane; MeOH, methanol.
Example 1
Figure imgf000054_0001
(D
Preparation of methyl 3-((2-oxo-2-(2-oxo-2,3-dihydro-l//-indol-5- yl)ethyl)thio)propanoate (I)
(i) 5-Bromoacetyloxindole (US 5849710) To a suspension of anhydrous aluminium, chloride {1 1,4 g, 85 nixnol) in 1 ,2-dichloroethane (25 ml) stirred at 0 PC was added bromoacetyl bromide (5.9 ml, 68 nimol) dropwise. After 1 h a suspension of oxindole (4.52 g, 34 mmol) in 1,2-dichlorøelhane (25 ml) was added and stirring continued for 2 h ai 0 DC then for 3 h at 50 0C. The reaction mixture was cooled, poured onio ice/water (500 nil) and filtered to give 5-broinoaeetylαxiπdole as a light brown solid (7.1 g, 82%) that was used without further purification.
(ϋ) Methyl 3-((2-σxo-2-(2-oxo-2,3-dihydro-l/f-mdol-5-yl)ethyl)thio)propanoatc (1)
To a suspension of 5-bromoacetyloxiadolc (4.15 g, 16.3 raraol) in N,N-dimethylforrnamide (20 ml) was added methyl 3-murcaptopropionate (2.14 ml, 19.6 mmol) and di- isopropylethylamine (6.1 ml, 35 mraol) resulting in a dark brown solution. The solution was stirred for 36 h at room temperature under an atmosphere of nitrogen then concentrated to give a yellow gum. Column chroinatography (SiO2) duting with 20: 1 chlorofbππ/MeOH afforded a dark yel low compound that was recrystallised from methanol to give the ester ( 1 ) as a light yellow solid (3.3 g, 72%), mp. 106-108 0C (TIX: RF = 0.64 on SiO2 with 9:1 dhlorofoπn/MeOH).
1I I nmr (300 MHa, d6-Jmso) 52.61, t (6.4 Hz), CH2; 2.70, t (5.8 Hz), CfI2; 3.54, s, H3; 3.57, s, OMc; 3.95, s, SCH2CO; 6.89, d (8.1 Hz)1 117: 7.81, s, H4; 7.87, br d (8.4 Hz), H6; 10.74, br s, Ni l,
ESI (+VO)" mfz 316 (M+Na, 20%), 294 (M+l I, 100%).
Example 2
Figure imgf000056_0001
(2)
Preparation of 3-((2-oxo-2-(2-oxυ-2r3-dihydrc)-l//-iαdoU5-yI)ethyI)thi<»)propajtioic acid (2)
A solution of the methyl ester (1) (3.0 g, 10,2 imnol) in concentrated hydrochloric acid (30 ml) was heated to reflux for 5 min then couletl to room temperature to give a yellow precipitate. The solid was filtered, washed with water and rccrystajlised from methanol to give the acid (2) as a light yellow solid (1.50 g, 52%), mp. 182-184 °C (TLC: Rκ = 0.31 on SiO2 with 9: 1 chlorofonπ/MeOH).
1I-I nmr (300 Ml Iz, oVdtnso) δ 2.5, obscured, CH2; 2.65, t (7.1 Hz), Cl I2; 3.54, s, 113; 3.94, s, SCH2CO; 6.89, d (8.1 Hx), 117; 7.82, s, H4; 7.87, d (8.4 Hz), H6; 10.74, br s, NH; 12.21, br ft, COOH.
ESI (+vβ) m/z 280 (M+H, 100%). ESI (-ve) m/z 278 (M-H, 100%).
Example 3
Figure imgf000057_0001
(3)
Preparation of methyl 3-((2-oxo-2-(2-oxo-2^3-dihydro-liy-benzimidaz;o]-5' yl)ethyl)thio)propaπιmte (3)
(i) 5-(Chloroacetyl)-1,3-dihydro-2W-ben7.imi<la7.ol-2-OLie (WO 92/50070)
Anhydrous aluminium chloride (7.5 g, 60 miπol) was crushed to a powder under nitrogen then suspended in 1 ,2-dichloroethane (10 ml). The suspension was cooled to 0 0C and chloroacetyl chloride (3.6 nil, 45 mmol) added dropwise. After stirring at 0 0C for 30 rain 2-hydroxybεπ7.imida7.ole (3.0 g» 22,4 mmol) was added portion-wise with additional 1,2- dichlorocihanc (5 ml). The reaclion mixture was heated For 2 h at 50-55 UC under nitrogen with vigorous stirring during which time the green-blue suspension became a dark solution. Afler stiiring for 16 h at room temperature lhe mixture was poured onto ice (100 g) and the resulting grey precipitate filtered. The solid was washed with water then ethyl acetate and dried under vacuum to give 5-(chloroacetyl)- 1 ,3-dihydro-2H-benzimidazol-2-onc as a light grey powder (4.7 g, 100%) that was used without further purification.
(ii) Methyl 3-((2-oxo-2-(2-oxo-2,3-dihydiO-l/f-bcnzimidazol-5-yl)elhyl)thio)propanoate (3)
To a solution of methyl 3-mercaptopropionate (0.57 g, 4.7 mmol) in dry tctrahydrofuran (15 ml) was added 5-(chloroacetyl)-l>3-dihydro-2/7-beπzimidaxoI-2-one (1.0 g, 4.7 mmol) followed by anhydrous potassium carbonate (3.3 g, 23.9 inrnol, 5 eq) and the mixture stirred at room temperature for 24 h. The reaction mixture was partitioned between ethyl acetate (50 ml) and water (50 ml) and the aqueous layer re-extracted with fresh ethyl acetate (50 ml). The combined organic extract was then washed with water (2 x 50 ml), brine (1 x 50 ml), dried (MgSO4) and the solution concentrated by rotary evaporator. Reducing the volume to 20-30 ml resulted in the formation of a precipitate which after chilling in ice was filtered to give the ester (3) as a red-brown solid (0.959 g, 69%), mp. 185-187 0C (TLC: R1. = 0.47 on SiO2 with 17:3 DCM/McOH).
1H nrnr (300 MHz, dή-dmso) δ 2.62, m, CH2; 2.72, m, CH2; 3.58, s, OMc; 3.99, a, SCH2CO; 7.00, d (8.1 HJH), 117; 7.48, d (1.5 Hz), H4; 7.67, dd (1.5, 8.1 Hz), H6; 10.86, s, NH; 1 1.03, s, NH.
ESr (+ve) m/z 317 (M+Na, 15%), 295 (M+H, 100%). ESI (-vβ) m/z 293 (M-I I, 100%).
Example 4
Figure imgf000058_0001
(4)
Preparation of 3-((2-oxo-2-(2'Oxo-2:3»dihydro-l/f-ben-dinidaizol-5- yl)ethyl)thiø)propanoic acid (4)
To a solution of the methyl ester (3) (300 mg, 1.02 mmol) in methanol (75 ml) was added 1 M sodium hydroxide solution (25 ml) and the solution stirred at room temperature for 4 h. The bulk of the methanol was then removed by rotary evaporator and the aqueous solution acidified with IM hydrochloric acid solution (25 ml). The cloudy suspension was then extracted with ethyl acetate (3 x 50 ml), the extract washed with brine (1 x 100 ml), dried (MgSO4) and evaporated to give the acid (4) as a pale yellow powder (0.229 gr 80%), mp. 212-215.°C (TLC: RH = 0.09 on SiO2 with ] 7:3 JDCM/McOH). 1H nmr (300 MHK, do-dmso) δ 2.53, t (6.9 Hz), CH2; 2.68, t (6.9 Hz), CH2; 3.98, s, SCH2CO; 7.00, d (8.1 Hz), H7; 7.48, d (1.2 Hz), H4; 7.67, dd (1.8, 8.1 Hz), Hό; 10.86, s, NH; 11-02, 8, NH; 12.21, br s, COOH.
ESI C+ve) m/t 303 (M+Na, 70%), 281 (M+H, 100%). ESI (-ve) mlz.219 (M-H, 100%).
Example 5
Figure imgf000059_0001
(5)
Preparation of methyl ((2-oxo-2-(2-oxo-2^-dihydro-lff-benzimidazoI-5- yl)ethyl)thio)acetate (5)
To a solution of methyl thioglycolatc (0.426 g, 4.01 minol) in dry tetrahydrofuran (30 ml) was added 5-(ch(oroacctyl)-l,3-dihydro-2//-bcnzitnidazol-2-one (0.80 g, 3.8 mmol) from example 3, followed by anhydrous potassium caτbonate (2.62 g, 5 eq). The mixture was stirred at room temperature for 90 h then partitioned between ethyl acetate (100 ml) and water (100 ml) and the aqueous layer extracted further with ethyl acetate (1x100 ml). The combined organic extract was washed with water (1 x 100 ml), brine (Ix 100 ml), dried (MgSO4) and the volume reduced by rotary evaporator to 30-40 ml. Reduction of the volume afforded a solid that was filtered off and dried under vacuum to give the ester (5) as a light orange powder (0.781 g, 73%), rap. 177-178.5 0C (TLC: Rp = 0.50 on silica gel with 17:3 DCM/MeOH).
1H nmr (300 MHz, dβ-dmso) 53.40, s, 0OCCH2S; 3.61, s, OMe; 4.11, s, SCH2CO; 7.01, d (8.1 I Iz), H7; 7.47, app s, H4; 7.66, dd (1.3, 8.1), H6; 10.87, s, NH; 11.04, s, NH.
ESI (+ve) m/z 303 (M+Na, 27%), 281 (M+H, 100%). ESI (-ve) m/z. T19 (M-H, 100%).
Figure imgf000060_0001
(6)
Preparation of ((2-c»x«.2.(2-oxo-2,3-dihydro-lW-benziniidiazoI-5-yI)ethyl)tliio)acetic acid (6)
To a solution of the tnclhyl ester (5) (0.30 g, 1.07 mmol) in methanol (75 ml) was added IM sodium hydroxide solution (25 ml) and the mixture stirred at room Lcmperaiure for 3.5 h. The bulk, of the methanol was then removed and the remaining aqueous solution acidified with IM hydrochloric acid (25 ml) while Stirling at 0 0C. n-Bitlanol (50 ml) and brine (50 nil) were then added and the aqueous layer re-extracted with n-butano) (50 ml). The • combined organic extract was washed with water (1x100 ml), brine (1x100 ml), dried (MgSO/i) and evaporated to give the acid (6) as an olive-green powder (0.238 g, 84%) which was recrystallised from methanol, mp. 230 0C (dec).
1H rimτ (300 MHz, dft-dmso) δ 3.24, s, 0OCCH2S; 4.06, s, SCH2CO; 7.00 d (8.1 Hz), H7; 7.48, d (1.5 Hz), 7.67, dd (1.5, 8.1 Hz), H6; 10.89, s, NH; 11.06, s, NH.
ESI (+vβ) m/z 3H (M+2Na-H, 20%), 289 (M+Na, 45%), 267 (M+H, 55%). ESI (-vc) m/z 287 (M+Na-2H, 20%), 265 (M-H5 100%). Example 7
Figure imgf000061_0001
(7)
Preparation of S-(((2-hydroxyethyl)thio)acetyl)-l,3-dihydiro-2i/-beiLziiiiida!«oI-2'one (7)
To a solution of 2-mercaptoethanol (0.30 g, 3.8 mniol) in dry tetrahydrofuraπ (30 ml) was added 5-(ch]oroacetyl)-l,3-dihydro-2/y-benziinidazol-2-one (0.80 g, 3.8 mmol) from exainple 3, followed by anhydrous potassium carbonate (2.62 g, 5 eq). The mixture was stirred al room temperature for 18 h then the rcaclitm mixture partitioned between elhyl acetate (100 nil) and water (100 ml) and the aqueous layer further extracted with ethyl acetate (1 x 100 ml). The combined organic extract was washed with water ( 1x100 ml), brine (1 x 100 ml), dried (MgSO4) and the volume reduced to 30-40 ml. Reduction of the volume afforded a precipitate that was filtered off and dried under vacuum to give the alcohol (7) as a light olive-green powder (0.285 g, 30%), mp. 320 0C (dec) (TLC: RF = 0.31. on SiO2 wilh 17:3 DCM/MeOH).
1H nmr (300 MHz1 dθ-dmso) δ 2.58, t (6.8 Hz), CH2S; 3.51, dt (5.7, 6.6 Hz), HOCH2; 3.95, s, SCH2CO; 4.74, t (5.4 Hz)5 HO; 7.00, d (8.1 Hz), 117; 7.48, d (1.2 Hz), H4; 7.66, dd (1.6, 8.2 Hz), H6; 10.86, br s, NH; 11.02, br s, NH.
ESI (+ve) m/z 275 (M+Na, 45%), 253 (M+H, 100%). ESI (-ve) m/z 251 (M-H, 100%). Example 8
Figure imgf000062_0001
(8) Preparation oF 6-({2-oxo-2-(2-oxo-2β-dihydro-l//-ii«loI-S-y[)ethj'I)thio)hexyl acetate (8)
A suspension of sodium hydride (0.237 g, 60% dispersion, 5.92 nυnol) in anhydrous N, N- dimethylformarnide (7 ml) was stirred at 00C for 5 min under nitrogen. 6-Mercapto-l- heλanol (0.059 ml, 0.43 minol) was added and storing continued at 0 "C for 20 min then 5- chloroacetyloxindole (0.099 g, 0.474 inrnol) was added and stirring continued at 0 "C for a further 1 h. The suspension was then partitioned between ethyl acetate and water and the aqueous phase acidified with IM hydrochloric acid and extracted with ethyl acetate. The combined organic phases were washed with IM hydrochloric acid, water, brine, dried (MgSO4) and concentrated to give a sticky yellow solid (0.231 g). Purification by column, chromatography (SiO2) elating with 99: 1 DCM/MeOH afforded the ester (8) as a white solid (0.085g, 51%), mp. 86-87 "C (TLC: RF = 0.44 on SiO2 with 9: \ DCM/McOH).
1H nmr (300 MHU, CDCl3) 6 1.38, m, 2xCH2; 1.57, m, 2xCH2; 2.04, s, Me; 2.57, t (7.2 Hz), CH2S; 3.60, s, H3; 3.73, s, SCH2CO; 4.04, t (6.9 Hz), OCH2; 6.92, d (8,1 Hz), H7; 7.69, br s, NH; 7.89, br s, H4; 7.93, br d (8.4 Hz), H6.
EST (+ve) m/z 372 (M+Na, 20%), 350 (M+H, 100%). EST (-ve) rn/z 348 (M-H, 10%). Example 9
Figure imgf000063_0001
(9)
Preparation of 5-(((6-hydroxyhexyl)thio)acetyI)-l,3-dihydro-2//-indoI-2-one (9)
After elutiofi with 99:1 DCM/MeOH as described in eΛample 8, further etution with 95:5 DCM/MeOH afforded the alcoliol (9) as a pale beige solid (0.048 g, 30%), mp. 105-1080C (TLC:. RF = 0.35 on SiO2 with 9:1 DCM/MeOH).
1H mar (300 MHz, dή-dmso) δ 1.30 - 1.55, m, 4xCH2; 2,4, obscured, CH2S; 3.35, dt (5.1 , 6.4 Hz). OCH2; 3.54, s, H3; 3.87, s, SCH2CO; 4.28, t (5.2 Hz), OH; 6.89, d (8.1 Rc), H7; 7.81, br s, H4; 7.87, dd (1.5, 8.3 Hz), H6; 10.73, s, NR
ESI <+ve) rn/z 330 (M+Na, 25%), 308 (M+H, 70%). ESI (-ve) m/z 306 (M-H, 60%).
Example 10
Figure imgf000063_0002
(10)
Preparation of 5-(((6-hydroxyhexyI)thio)acety1)-l^-dihydro-2//-beπziraidazoI-2-one (10) To a solution of 6-mercapto- I.-hexanol (0.51 g, 3.8 mmol) in tetrahydroruran (30 ml) was added 5-(chlcjroacetyl)-i,3-dihydτo-2Λf-benziEαidazol-2-one (0,80 g, 3.8 mmol) from example 3, followed by dry potassium carbonate (2.62 g, 19.0 mmol, 5 eq) and the suspension stirred at room temperature for 96 tu The reaction mixture was partitioned between waler (100 ml) and ethyl acetate (80 ml) and the aqueous layer re-ex trøctcd with ethyl acetate (80 ml). The combined organic extract was washed with water (1 x 100 ml), brine (1 x 100 m!), dried (MgSO4) and concentrated Io a volume of 30-40 ml resulting in precipitation. The precipitate was filtered off to give the alcohol (10) as a pale yellow powder (0.658 g, 56%), mp. 180-181 "C.
1H πmr (300 MH-:, d^-dmso) 6 1.2-1.55, m, 4 x CH2; 2.5, obscured, CHiS; 3.35, t (6.5 Hz), OCH2; 3.89, s, SCH2CO; 4.2, br s, OH; 6.99, d (8.1 Hz), H7; 7.48, d (1.2 Hz), H4; 7.66f dd (1.6, 8.2 Hz)7 H6; 10.85, s, NH; 1.1,02, S, NH.
ESt (+ve) tnfz 331 (M+Na, 40%), 309 (M+U, 100%). EST (-ve) m/z 307 (M-H, 100%).
Example 11
Figure imgf000064_0001
(ID
Preparation of 6-chIyro-5-(((6-hydroxyhexyI)thio)acetyl)-13-dihydn»-2W-indol-2-one (H)
A suspension of 5-chloroacctyl-6-chIorooxindolc (0.099 g, 0.407 rnmol), 6-mercaρlo-l- hexanol (0,062 ml, 0.453 mmol) and potassium carbonate (O.059 g, 0.43 mmol) in acetonitrile was heated at τeflux under nitrogen for 2.5 h then cooled to room temperature. Tlic suspension was filtered and the filtrate concentrated to give a dark red-brown solid (0.163 a). The solid was ourified bv column chromatoeraDhv CSiO?) ehitinε with 100% DCM, 99:1 and 95:5 DCM/MeOH to give the alcohol (11) as a pale yellow solid (0.091g, 65%), rap. 106-108 0C (TLC: RH = 0.42 on SiOj with 9: 1 DCMMeOH).
Ηnmr (3CK) MHz, CDCl3) 6 1.38, m, 2xCH2; 1.53, m, 2χCH2; 2.54, br s, CH2S; 3.56, s, H3; 3.64, t (6.3 Hz), OCH2; 3.84, br a. SCl I2CO; 6.95, s, 117; 7.52, s, H4; 8.63, s, NH,
ESI (+vβ) iti/z 364/366 (M+Na, 25/8%), 342/344 (M+H, 100/30%). ESI (-ve) m/z 340/342 (M-] MOO/35%).
Example 12
Figure imgf000065_0001
(12)
Preparation of methyl 3-«2-(6-chloro-2^>xo-2»3-dihydro-l/J-indol-S-yl)-2- oxoethyl)thio)propanoate (12)
A suspension of 5-chloroacet;yl-ό-chloiOθxindo[e (0.100 g, 0.413 nuαol), methyl 3- mercaptopropionaie (0.050 ml, 0.45 mmol) and potassium carbonate (0.057 g, 0.41 mπiol) in acetonittile (3 nil) was refluxed under nitrogen for 21) then cooled to room temperature. The suspension was filtered, washing with dichloromethanc and the combined filtrate and washings concentrated to give a red-brown solid (0.147 g). The solid was purified by column, chromatography (S1O2) eluting with 100% DCM and 99:1 DCM/MeOH to give the methyl ester (12) as a beige solid (0. ! 07 g, 79 %)t mp. 75-7 0C (TLC: RF = 0.20 on SiO1 with 95:5 DCMMeOH).
1H nmr (300 MHz, 4-dmso) δ 2.61, m, Clhl 2.70, m, CH2; 3.52, β, H3; 3.58, s, OMe; 3.93, s, SCH2CO; 6.88, s, H7; 7.67, s, H4; 10.73, s, COOH. ESl (+ve) m/z 350/352 (M+Na, 90/30%), 328/330 (M+H, 100/30%). ESI (-ve) mk 326/328 (M-H, 30/10%).
Example 13
Figure imgf000066_0001
(13)
Preparation of S-tø-Cή-chlorø^-oxo^^-dihydro-lff *indoI-5-yl)-2- ox(K!thyJ)thio)projianoic acid (13) (Method 1)
The methyl ester (12) (0.05 J g, O. ld mmol) was treated with concentrated hydrochloric acid (2 itil) and heated at reflux briefly (< I niiα) then cooled to room temperature. The suspension was filtered, the solid washed carefully with, water and dried under vacuum to give the acid (13) as a light brown solid (0.041 g, 83 %), nip. 203-5 0C (TLC: Rf = 0.63 on SiO2 with 8:2 DCM/McOH).
1H πmr (300 MHz, d(,-dniso) 62.5, obscured, CH2; 2.66, t (6.6 Hz), CH2; 3.53, s, H3; 3.92, s, SCII2CO; 6.88, s, H7; 7.67, s, H4; 10.73, s, NH; 12.23, br s, COOH.
ESI (+ve) mfz 336/338 (M+Na, 10/4%), 314/316 (M+H, 15/4%). EST (-vc) wt 312/314 (M-H, 100/35%).
Preparation of 3-((2-(<J-chIoro-2-oxo-2»3-dihydro-ll-'-mdol-5-yl)-2- oxouthyl)thio)propaαoic acid (13) (Method 2)
5-Chloroacctyi-6-chlorooxindo]e (1.2 g, 4.8 mmol), 3-mercaρtoproρionic acid (0.60 g, 0.5 ml, 5.65 mmol) and DMF (5 nil) were added to a 50 nil flask. Diisopropylethylaminc (1.8 ml, 10.3 mmol) was added to the reaction mixture with stirring which was continued for 10 h at room temperature under nitrogen. The reaction mixture was then added dropwise with stirring to 200 nil of 10% citric acid solution resulting in the formation of a white precipitate. After cooling in a refrigerator for 4h the solid material was filtered, washed with water (3x50 ml) and hexane (3x20 nil) then dried under vacuum to give the acid (13) as an off- white solid (1.43 g, 95%), identical to the material prepared by Method 1.
Using Method 2 described above, examples 14-21 were prepared by reaction of either 5- chloroaceiyloxuidole, 5-chlon>acetyl-6-chloroox indole, 6-chloroacetyl-2-beiizoxazolinone or ό-bromoacetyl-Z-benzothiazoIinone, with 3-inercaptoρropioftie acid, 6-inercapto-l - hexanol, 1-bυtaαethiol or thioglycolic acid.
Example 14
Figure imgf000067_0001
(14)
3-((2-Oxo-2-(2»αxo-2?3-dihydrft- 1.3-bertzoxazol-ό-yr)ethy l)thio)ρropauoic acid (14)
1H nmr (400 MHz, d^-dmso) 52.46 (L, 2H); 2.61 (t, 2H); 3.95 (s, 2H); 7.1 S (d, IH); 7.82 (m, 2H).
Example 15
Figure imgf000068_0001
(15)
6-(((6-HydroxyhexyI)thio)acetyϊ)" l,3-beαzoxaz;ol-2(3J--0.»πc (15)
1H nmr (400 MHz, dή-dmso) δ 1.1 -1.5 (ni, 8H); 2.42 (m, 4H); 3.89 (β, 2H); 4.3 (t, IH); 7.15 (d, IH); 7.8 (m, 2H); 12. Ks, IH).
Example 16
Figure imgf000068_0002
(16)
6-((Butylthio)acetyI)-l,3-benzoxa5;ol-2(3i/)-one (16)
1H nmr (400 MH?., d^-dniso) δ 0.79 (ι, 3H); 1.28 (in, 2H); 1.42 (m, 2H); 2.42 (m, 211); 3.9 (s, 2H); 7.18 (d, IH); 7.85 (m, 2H). Example 17
Figure imgf000069_0001
(17)
((2-Ox«>-2-(2.oxo-2,3-diliydro-l^-ben-!;oχa-ci)l«6-yl)ethyl)thio)acetic acid (17)
1H. iimr (400 MHz, de-dmso) δ 3.2 (s, 2U); 4.05 (s, 2H); 7.15 (d, IH); 7.8 <τn, 2H).
Example 18
Figure imgf000069_0002
(18)
3-((2-Oxo-2-(2-oxo-2t3-dihj'dro. ϊ,3-bcnzothiazol-6-yI)ethyl)thi«)propanoic add (IS)
1H mar (400 MHz, dn-dmso) B 2A' b (m, 2H); 2.60 (m, 211); 3.95 (s, 2H); 7.15 (d, IH); 7.86 (d, I H); 8.23 (s, IH); 12.27 (s, IH). Example 19
Figure imgf000070_0001
(19)
Λ-({ButyIthio)acetyI)-l,3-bcπ/othiazpI-2(3fl)-one (19)
1H nmr (400 MHA 4-dmso) δ 0.79 (rn, 311); 1-26 (m, 2H); 1.43 (m, 2H); 2.42 (m, 2M); 3.87 (s, 2H); 7.1.5 (d, 1 H); 7.86 (m, IH); 8.22 (s, IH); 12.27 <s, IH).
Example 20
Figure imgf000070_0002
(20)
5-((Butylthio)acetyl)-6-cliloro-l3-dihydro-2ff-indol-2-one(20)
1H nmr (400 MHz, de-dmso) δ 0.79 (l, 3H); 1.25 (m, 2H); 1.42 (m, 2H); 2.42 (ι, 2H); 3.49 (s, 2H); 3,81 (s, 2H); 6.84 (s, IH); 7.63 (s, 111); 10.74 (s, IH). Example 21
Figure imgf000071_0001
(21)
5-((Butylthio)acetyl)-13-dihydro-2//-indol-2-one (21)
1H nmr (400 MHz, ds-dmso) δ 0.79 (t, 3H); 1.25 (m, 2H); 1.43 (m, 2H); 2.42 (l, 2H): 3.50 (s, 2H); 3.83 (B, 2H); 6.85 (d, IH); 7.77 (s, IH); 7.83 (d, IH); 10.75 (s, IH).
Example 22
Figure imgf000071_0002
(22)
Preparation of 5-((butylthio)acetyl)-l^-dihydro-2//-bcnzimidazoI-2-one (22)
1-Butanethiol (647 mg, 7.17 mmol) was dissolved in anhydrous THF (24 ml) and 5- (chloroacetyl)-l,3-dihydro-2//-beniώnidazol-2-onc (see example 3) (1.497 g, 7.11 mmol) and anhydrous potassium carbonate (4.938 g, 35.7 mmol) were added. The mixture was stirred at room temperature overnight then the reaction mixture was partitioned between ethyl acetate (75 ml) and water (75 ml). The aqueous phase was extracted with ethyl acetate (75 ml) and the combined ethyl acetate extracts were washed with water (2 x 75 ml) and brine (75 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated to appro* 30 ml and chilled overnight. The mixture was then filtered and the residue dried under vacuum to give the title compound ( 1.429 g, 76% yield) as a brown powder, mp 211-213 0C.
1H nmr (400 MI ϊz, d6-dmso) 6 0.85 (t, J = 7.4Hz, 3H); 1,32 (sextet, J = 7.5H-;, 2H); 1.50 (quintet, / = 7.3Hz, 2H); 2.47-2.53 (resonance obscured by residual d5-dmso); 3.92 (s, 2H); 7.02 (d, J = 8.4Hz, IH); 7.50 (d, J = 1.6Hz, IH); 7.68 (dd, / = 8.2, 1.8Hz, I H); 10,90 (br s, IH); 1 1.07 (br s, IH).
Example 23
Figure imgf000072_0001
(23)
Preparation of 3-((2-(l,3-dimethyl-2-oxu-2)3-dihydro-l//-benzimidazol-5-yl)-2- oxoethyl)tbio) propanoic acid (23)
(i) l,3-Dimethyl-l,3-dihydro-2W-bcn7.iiτiidazol-2-onc
l,3-Dihydro-2H-benziiuida7;ol-2-one (7.522 g, 56.1 mmol) was dissolved in anhydrous DIvIF (125 oil) and anhydrous potassium carbonate (46.581 g, 337 mmol) and iodomethane (21 ml, 337 mmol) were added then the mixture stirred at room temperature overnight. The reaction mixture was poured into chloroform (500 ml), filtered and the filtrate was evaporated to dryness. The resultant residue was dissolved in a mixture of ethyl acetate (150 ml) and water (100 ml). The cthyi acetate phase was washed with water (2 x 100 ml) and brine ( 100 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (7,229 g, 79% yield) as a pale yellow solid.
1H nmr (400 MHz1 CDCl3) δ 3.43 (s, 6H); 6.95-7.01 (m, 2H); 7.08-7.14 (m, 211). (ii) 5-(Chloroacctyl)-l,3-climethyl-l,3-dihydro-2//-beπ7,imida_;ol-2One
Aluminium chloride (13.427 g, 101 mmol) was suspended in DCE (80 ml), cooled in an ice bath and chloroacctyl chloride (6.4 ml, 80 mmol) added dropwi.se with a glass dropping pipette. The mixture was stirred at O0C under nitrogen for 30min then 1 ,3-diinethyl- 1 ,3- dihydro-2tf-beimrnidazol-2-c>πe (6.504 g, 40-1 mmol) was added in portions. The mixture was healed at 550C under nitrogen for 2h, then allowed to cool to room temperature and poured onto ice (200 g). The mixture was filtered and the residue washed with water (100 ml). The filtrate contained two phases which were separated. An attempt was made to dissolve the residue in a mixture of the DCE phase from the filtrate, additional DCH (50 ml) and chloroform (150 ml). The residue only partially dissolved and washing this mixture with water ( I (X) ml) gave an emulsion. The mixture in the separating funnel was filtered and the remaining solid in the separating funnel was suspended in water (3 x 100 ml) and ethyl acetate (40 ml). Each of the washes was filtered and the residue was dried at the pump and then dried under vacuum over silica gel overnight to give the title compound (7.003 g, 85% yield) as a pink solid.
1H nmr (400 MHz, d<-dmso) S 3.36-3.41 (m, 6H); 5.18 (s, 2H); 7.30 (d, J = 8.4Hz, IH); 7.76 (d, / = .1.2Hz, IH); 7.81 (dd, /= 8.2, 1.4Hz, IH).
(iπ) 3-((2-(l,3-Dimethyl-2-oxo-2,3-dihydro-lH-bcnzimid-ιzol-5-yl)-2- oxoelhyl)thio)propan.oic acid (23)
3-Merca.ptopropionic acid (583 mg, 5.49 mmol) was dissolved in anhydrous DMF (17 ml)
Figure imgf000073_0001
mmol) and anhydrous potassium carbonate (3.796 g, 27.5 mmol) were added. The mixture was stirred under nitrogen for 75min then the reaction mixture was partitioned between ethyl acetate (150 ml) and hydrochloric acid (IM, 80 ml). The aqueous phase was extracted with ethyl acetate ( 100 ml) and the combined ethyl acetate extracts were washed with water (2 x 100 ml) and brine (100 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (1.149 g, 69% yield) as a pale orange solid, mp 174-176 0C. 1H nmr (400 MHz, dό-dmso) 8 2.54 (l, J = 7.2Hz, 2H); 2.70 (t, J - 7.0Hz, 2H); 3.38 (s, 3H); 339 (s, 3H); 4,05 (s, 2H); 7.26 (d, J = 8.0Hz, IH); 7.76 (d, J = 1.6Hz, IH); 7.81 (dd, J= 8.2, 1.8Hz, IH); 12.28 (br s, IH).
Example 24
Figure imgf000074_0001
(24)
Preparation of 5-((but3'lthio)acetyl)-l,3-dimethyl-l!3-dihj'dπ»-2/f-biinϊ!imidazoJ-2-one
1-Butanethiol (616 mg, 6.83 ininol) was dissolved in anhydrous THF (21 nil) and 5- (chloroacetyl)-l ,3-dimethyl- 1 ,3-dihydro-2/f-benzirnidazol-2-one (1.604 g, 6.72 mvπol) and auliydrόus potassium carbonaic (4.633 g, 33.5 mmol) were added. The mixture was stirred at room temperature for 4 days before the reaction mixture was partitioned between ethyl • acetate (60 ml) and water (60 ml). The aqueous phase was extracted with ethyl acetate (60 ml) and the combined ethyl acetate extracts were washed with water (2 X 60 ml) and brine (60 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give an orange oil which solidified on standing. The solid was broken up to give the title compound (1.868 g, 95% yield) as a yellow powder, mp 80-81 0C.
1H tarn (400 MHz, d^dmso) δ 0.86 (t, J = 6.8Hz, 3H); 1 ,32 (sextet, J = 7.3Hz, 2H); 1.51 (quintet, J = 7.3Hz, 2H); 2.49-2.54 (resonance obscured by residua! d5-dmso); 3.37 (s, 3H); 3.39 (s, 3H); 3.98 (s, 2H); 7.25 (d, J = 8.4Hz, IH); 7.75 (d, J= 1.6Hz, IH); 7.81 (dd, J = 8.2, 1.4Hz, IH). Example 25
Figure imgf000075_0001
Preparation of 5-((butylthio)acetyI)-6-chloro- j^-dϋiydro-iff-benzimidazol^-oiie (25)
(i) 5-Chloro-6-(chloroaceLyl)-l,3-clihydiO-2//-bcπzimidazol-2-one
Aluminium chloride (9.953 g, 74.6 mmol) was suspended in DCE (20 ml) and cooled in an ice bath. Chloroacetyl chloride (4.70 nil, 59.0 mmol) was added dropwise with a glass dropping pipette and the mixture was stiired at O0C under nitrogen for 30min. 5-Chloro- l,3-dihydro-2/J-bcnzimidazol-2-one (5.0(K) g, 29.7 mmol) was added in portions and the mixture was heated at 55"C under nitrogen for 3Vih. The mixture was allowed to stand at room temperature under nitrogen overnight, lhen heated at 55°C under nitrogen for a further 5VJII. The reaction mixture was allowed to cool Io room temperature and poured onto ice (400 g) and filtered. The residue was washed with water (2 x 100 ml) and dried at the pump. The residue was washed with ethyl acetate (20 ml, 3 x 40ml) and dried at the pump to give the title compound (2.631 g, 36% yield) as a dark green powder. The product contained 10 mol% 5-chloro-l,3-dihydro-2H-bcnzimidazol-2-oήe.
1H mXiT (400 MHR, dβ-dmso) δ 5.04 (s, 2H); 7.06 (s, IH); 7.36 (s, IH); 11.1 1 (br s, IiI); 11.15 (br s, I H).
(ii) 5-((ButyIthio)acetyl)-6-chloru-l,3-dihydro-2//-benziniida'-:ol-2-ore (25)
1 -Butanethiol (478 mg, 5.30 mmol) was dissolved in anhydrous DlVfF (17 ml) and 5-chloro- 6-(ch)oroacctyl)-l ,3-dJhydro-2//-benzimida7.ol-2-one ( 1.297 g, 5,29 mmol) and anhydrous potassium carbonate (3.666 g, 26.5 mmol) were added. The mixture was stirred under nitrogen for 4OmIu then the reaction mixture was partitioned between ethyl acetate (100 ml) and hydrochloric acid (3M, 80 ml). The aqueous pliase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (50 ml). A further portion of ethyl acetate (100 ml) was added to lhe ethyl acetate phase and the ethyl acetate extracts were washed with water (50 ml) and brine (50 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give a red grey powder (1.441 g, 91% yield). 1H nmr analysis showed that the product contained 10 mol% unchanged starting material. The crude product was dissolved in anhydrous DMF (15 ml) and a. solution of 1-butanethiol (100 mg, 1.11 mniol) in anhydrous DMF (2 ml) was added followed by anhydrous potassium carbonate (759 mg, 5.49 mmol). The mixture was stirred under nitrogen for όOmin before the mixture was partitioned between ethyl acetate ( 100 ml) and water (100 ml). The aqueous phase was extracted with ethyl acetate (100 ml) and the combined ethyl acetate extracts were washed with water (2 x 75 ml) and brine (75 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give a brown solid which was transferred to a sinter funnel, washed with absolute ethanol (20 ml) and dried at die pump. The residue was dried under vacuum over potassium hydroxide pellets overnight to give the title compound (880 mg, 56% yield) as a pink, powder, mp 199-201 0C.
111 nmr (400 MHz, dV-drnso) δ 0.84 (t, J = 7.2Hz, 3H); 1.31 (sextet, J - 7.4Hz, 2H); 1.48 (quintet, / = 7.4Hz, 2H); 2.48 (resonance obscured by residual drdmso); 3.89 (s, 2H); 7.02 (s, I H): 7.30 (s, 1 H); 1 1.05 (br a, 2H).
Example 26
Figure imgf000077_0001
(26)
Preparation of 3-((2-(6-chloro-2-oxo-2r3-dihyrtro-l£r-benzimidazol-5-j'l)'2- oxouthyl)thio)propanoic acid (26)
3-Mercaptoρropionic acid (566 nig, 5.33 minol) was dissolved in anhydrous DMH" (17 ml) and 5-chloro-6~(chloroacctyl)-13-dihydra-2H-bcn7.imidazol-2-one (1.30U g, 5.30 nunol) and anhydrous potassium carbonate (3.714 g, 26.9 itunol) were added. The aύxture was stirred under nitrogen for 35min then the reaction mixture was partitioned between ethyl acetate (100 ml) and water (150 ml). Emulsions prevented the separation of the phases and hydrochloric acid (J M1 80 ml) was carefully added. The phases were separated and the aqueous-phase was extracted with ethyl acetate (100 ml, 50 ml). The combined ethyl acetate extracts were washed with water (2 x 100 ml) and brine (100 ml), dried over anhydrous magnesium sulfate, and filtered. The filtfaic was evaporated to dryness to give a dark green powder.
The crude product was partitioned between ethyl acetate (150 ml) and a 5% sodium hydrogen carbonate solution (200 ml). The ethyl acetate phase was extracted with water (100 ml) and the combined aqueous phases were washed with ethyl acetate (100 ml), acidified with hydrochloric acid (3M, 50 ml) and extracted with ethyl acetate (300 ml, 2 x 100 rnl). There was a significant quantity of a pale brown solid that did not dissolve. The aqueous phase containing the emulsion was filtered and dried at the pump to give the title compound (447 mg, 27% yield) as a cream solid. The ethyl acetate extracts were evaporated to dryness to give a green solid and the residue partitioned between ethyl acetate (ISO ml) and a 5% sodium hydrogen carbonate solution (200 ml). The aqueous phase was acidified with hydϊoclilorie acid (3M, 50 ml) and the resultant suspension washed with ethyl acetate (50 ml). The combined aqueous and ethyl acetate phases were filtered and the residue was washed with water (2 x 50 ml) and dried at the pump to give the title compound (579 mg, 35% yield) as a pale green solid.
The two batches of 3-((2-(6-chloro-2-oxo-2,3-dihydro-ΪH-benzimidazol-5-yl)-2- oxoethy])thio)propanoie acid were dried under vacuum over silica gel overnight Both samples had a similar appearance after drying and the two samples were combined to give the title compound (1.013 g, 61% yield) as a pale beige powder, mp 186-187 "C.
1H αmr (400 MHz, dg-dmso) δ 2.51 (resonance obscured by residual d5-dmso); 2.68 (I, / = 7.2Mz, 2H); 3.97 (s, 2H); 7.02 (s, IH); 7.31 (s, IH); 11.03 (br s, IH); 11.09 (br s. I H); 12,29 (br s, IH).
Example 27
Figure imgf000078_0001
(27)
Prcparatiort of 5-((butyIthio)acetyl)-6-chloro-l,3-dimethyl-l,3-dihydro-2f/- benzimidazol-2-one (27)
(i) 5-Chl«π>-ϊ ,3-dime-liyl-l,3-dihydrϋ-2H-beimaiidazo!-2-onc
5-Chloro-l ,3-dihydro-2/?-ben^imidazol-2-0Qc (7.040 g, 41.8 mmol) was dissolved in anhydrous DMF (100 ml), anhydrous potassium carbonate (34.707 g, 251 mmol) and iodomcthane (15.5 ml, 249 mmol) were added and the mixture stirred at room temperature overnight. The reaction mixture was poured into chloroform (400 ml) and mixed well then filtered and the filtrate evaporated to dryness. The resultant residue was dissolved in a mixture of ethyl acetate (500 in I) and water (200 ml), the ethyl acetate phase was washed with water (2 x 100 ml) and brine (100 ml), dried over anhydrous magnesium sulfate and filtered, The filtrate was evaporated to dryness to give the title compound (J.163 g, 87% yield) as a brown powder.
1H nmr (400 MH?-, CDCIa) δ 3.37-3.42 (m, 6H); 6.87 (d, J - 8.0Hz, IH); 6.97 (d, J = 1.6Hz, IJ I); 7.07 (dd, J = 8.2, 1.8Hz, IH).
(ii) 5-Chloro-6-(chloroat;etyl)- 13-dimet,hyl-l,3-dihydro-2//-benziiτiidazol-2-one
Aluminium chloride (8.589 g, 64.4 mmol) was suspended in DCE (17 nil) arid cooled in an ice bath. Chloroacetyl chloride (4.05 ml, 50.8 mmol) was added dropwise with a glass dropping pipette and the mixture was slirred at O0C under nitrogen for 30min. 5-Chloro- l,3-dimethyl-l,3-dihydro-2J/-benzimidazol-2-one (5.010 g, 25.5 mmol) was added in portions and Lhc mixture was heated at 55°C under nitrogen for 3h. The mixture was allowed to cool to room temperature and poured onto ice (200 g) then filtered. The residue was washed with water (3 x 100 ml), dried at the pump and then dried under vacuum over silica gel to give the title compound (5.573 g, 80% yield) as a brown powder.
1H nmr (400 MHz, ds-dmso) δ 3.33-3.37 (m, 6H); 5.09 (s, 2H); 7.45 (s, J H); 7.69 (s, IH).
(iii) 5-((Butylthio)acetyl)-6-chloro-l ,3-dimelhyl-l,3-dihydro-2H-benzimidazol-2-onc (27)
1-Butanetbiol (533 mg, 5.91 mmol) was dissolved in anhydrous THF (19 ml) and 5-chloro- 6-(chloroacetyl)-l,3-dirnethyl-l,3-dihydro-2rT-benziinτdazol-2-one (1.598 g, 5.85 mmol) and anhydrous potassium carbonate (4.071 g, 29.5 mmol) were added followed by anhydrous DMF (1.0 ml). The mixture was stirred at room temperature for 5 days then the reaction mixture was partitioned between ethyl acetate (60 ml) and water (60 ml). The aqueous phase was extracted with eihyl acetate (60 ml) and the ethyl acetate extracts were washed with water (2 x 60 ml) and brine (60 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness and the resultant residue purified by bulb-to-bulb distillation (2500C / 0.57mbar) to give the title compound (1.037 g, 54% yield) as a pale yellow solid, mp 66-68 0C,
1H nmr (400 MHz, drdmso) δ 0.85 (t, J = 7.4Hz, 3H); 1.32 (sextet, J = 7.3Hz, 210; 1-49 (quintet, /= 7.4Hz, 2H); 2.48-2.53 (resonance obscured by residual d5-dmso); 3.34-3.37 (to, 6H); 3.95 (s, 2H); 7.40 (s, IH); 7.62 (s, IH).
Example 28
Figure imgf000080_0001
(28)
Preparation ι>r3-((2-(6-chloro-l)3-diiin:thyl-2-ox*>-2^J-dihydro-l--?-ben-!imidaz4>I-5-yI)- 2-oxoethyl)thio)propanoic add (28)
3-Mcrcaptopropioπie acid (593 mg, 5.59 mmol) was dissolved in anhydrous DMF (17 ml) and 5-chloro-6-(chloi'oacetyl)-l,3-dimethyl-l,3-dihydro-2W-ben7.imida2ol-2-onc (1.498 g, 5.48 minol) and anhydrous potassium carbonate (3.784 g, 27.4 mmol) were added. The mixture was stirred under nitrogen for 35min then partitioned between ethyl acetate (100 nil) and hydrochloric acid ( 1 M, 80 ml). The aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 50 ml) and brine (50 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (1.797 g, 96% yield) as a bruwn powder, mp 148-150 0C.
1H nmr (400 MH?., dή-dmso) δ 2.53 (resonance obscured by residual d^-dmso); 2.69 (t, J - 7.2H7, 2H); 3.35 (s, 3H); 3.36 (s, 3H); 4.02 (s, 2H); 7.41 (s, IH); 7.63 (s, H O; 12.30 (br s, IH). Example 29
Figure imgf000081_0001
(29)
Preparation of 6-((butylthio)acetyI)-5-chIoro-l^-benzothiazol-2(3Jtf)-one (29)
(i) 5-Chloro-6-(chloroacetyl)- 1 ,3-beπz(jώiai:ol-2(3H)-one
Anhydrous DMF (8.2 ml) was added dropwise to aluminium chloride (40.846 g, 306 mmol) with stirring (Caution: exothermic). The mixture was stirred until an even slurry formed (.hen 5-chloro-2-benzothiazαlonc (7.020 g, 37.8 mmol) was added in portions. The mixture was heated at 7O0C under nitrogen then bromoacctyl bromide (5.6 ml, 64 mmol) was added and the mixture heated at 700C under nitrogen for 25h. The mixture was allowed to stand at room temperature under nitrogen overnight then was poured onto ice (200 g), slirred for Ih and filtered. The residue was washed with water (2 x 100 ml) and dried at the pump then washed with ethyl acetate (3 x 25 ml) and dried at the pump to give the title compound (4.779 g, 41% yield) as a dark green powder. 1H nmr analysis showed thai ihc product also contained 6-(brornoacccyl)-5-chloro- 1 ,3-beπzothiai_ol-2(3H)-one (23 mol%) and an unidentified impurity (14 axo\%).
1H nmr (400 MH/., df,-dmso) 8 5.04 (s, 2H); 7.22 (s, I H); 8.17 (a. IH); 12.41 (br s, IH). Bromoacetyl impurity: ό 4.84 (s, 2H); 7.22 (s, IFT); 8.19 (s, IH); 12.41 (br s, IH), Unidentified impurity: δ 7.27 (s, 1 H); 8.08 (s, IH); 12.17 (br S, 111).
(ii) 6-((Butylthio)acetyl)-5-chloro-l t3-benzothiazol'2(3H)-one (29)
1-Butanethiol (557 mg, 6.18 mmol) was dissolved in anhydrous DMF (19 ml). A mixture of 5-chloro-6-(chloπjacei.yl)-l,3-bcuzothiazol-2(3/:/)-one (63 mol%), 6-(bromoacetyl)-5- chloro- 13-benzothiazol-2(3H)-one (23 mol%) and an unidentified impurity (14 mol%) (1,610 g, 6.14 Ttirπol) and anhydrous potassium carbonate (4.236 g, 30,6 mmol) were added. The mixture was stirred at room temperature under nitrogen for 105 min then was partitioned between ethyl acetate ( 150 ml) and hydrochloric acid ( I M, 80 ml). The aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 75 ml) and brine (75 ml), dried over anhydrous magnesium sulfate, and filtered. The filtrate was evaporated to dryness and the resultant residue dried under high vacuum ( 1000C / 0.8mbar for 5min) Io give a dark brown solid (1.317 g). A portion of the crude product (325 mg) was dissolved in ethyl acetate (20 ml) and silica gel 60 (1.5 g) was added and the mixture evaporated io dryness and purified by flash chromatography over silica gel 60 (eluent: 30% ethyl acetate / petroleum spirits (5 x 20. ml fractions), packing height: 20 cm, column diameter: 1 cm for lS)cm, then 2.5cm), The fractions containing the fivst major band (R/0.40, clucnl: 30% ethyl acetate /petroleum spirits, fractions 2-4) were combined and evaporated to dryness. The residue was dried under high vacuum (100 0C / O.Smbar for 5min) to give the Lille compound (248 mg, 13% yield) as an orange melt, mp 102.5-115.0 UC. 1H nmr analysis showed that the product contained the unidentified impurity (20 mol%) that was present in the starting material.
1H nmr (400 MHz, dti-din.so) δ 0.85 (t, /= 7.4Hz, 3H); 1.31 (sextet, J = 7.41 iz, 2H); 1.48 (quintet, /= 7.4Hz, 2H); 2.49 (resonance obscured by residual d5-dtnso); 3.89 (s, 2H); 7.19 (s, IH); 8.13 (s, IH); 12.29 (br s, IH). Impurity present in starting material: δ 7.27 (s, IH); . 8.08 (s, IH).
Example 30
Figure imgf000083_0001
(30)
Preparation of S-^-CS-chloro-l-nxo^^-dihydro-lβ-bcπssothiazoI-β-j'l)^- oxoethyl)thio)propan<>ϊc acid (30)
(i) 6-(Bromoacctyl)-5-chloro-l,3-bciii.oLhiii7.o!-2(3//)-onc
Anhydrous DMF (8.2 nil) was added dropwise to aluminium chloride (40,556 g, 304 inmol) with stirring (Caution: exothermic). The mixture was stirred until an even slurry formed then 5-chloro-2-benzothiazolone (7.030 g, 37.9 mmol) was added in portions. The mixture was heated at 700C under nitrogen and bromoacetyl bromide (5.6 nil, 64 miriυl) added and the mixlure heated at 700C under nitrogen for 71ZJII. The mixture was allowed to stand at room temperature under nitrogen overnight then the :tnix.lure was poured onto ice (200 g), stirred for I h and filtered. The residue was washed with water (2 x 100 ml) and dried at the pump. The residue was then washed with ethyl acetate (2 x 40 ml) and dried at the pump to give the title compound (3.150 g, 27% yield) as a tan powder. 1I I ninr analysis showed that the product also contained 5-chloro-2-benzothiazυlι>ne (33 mol%) and 5-chloro-6- (chloroacctyl)-l,3-benzothiazol-2(3//)-one (32 mol%).
1H πmr (400 MHz, dβ-dmso) δ 4.84 (s, 2H); 7.22 (s, IH); 8.19 (s, IH); 12.41 (hr s, IH). Starting material: δ 7.12 (d, J = 2.0Hz, IH); 7.19 (dd, / = 8.4, 2.0HT:. IH); 7.62.(d, / = 8.4Hz, IH); 12.06 (br s, IH). Chlαroacetyi impurity; δ 5.04 (s, 2H); 7.22 (s, IH); 8.17 (s, IH); 12.41 (br s, IH). (ii) 3-((2-(5-chloro-2-oxo-2,3-tiihydro-i,3-beii2otliiazol-6-yl)-2-oxoethyl)thio)prϋpant)ic acid. (30)
3-Mercaptoproρionic acid (566 mg, 5.33 irnnol) was dissolved in anhydrous DMF (17 ml) and a mixture of 6-(broraoacctyl)-5-chloro-l ,3-benzotliiazol-2(3fiO-one.(3S mol%), 5- chloro-ό^chloroacelyl)-! 3-benzothiazol-2(3H)-oπe (32 mol%>) and 5-ehloro-2- benzothiazoione (33 mol%) (1.999 g, 5.31 mmol based on available bromoacetyl and chloroaceiyl compounds) and anhydrous potassium carbonate (3.707 g, 26.8 mmol) were added. The mixture was stirred under nitrogen for 45min then the reaction mixture was partitioned between ethyl acetate (150 ml) and hydrochloric acid (IM, 80 ml). The aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 100 ml) and extracted with a 5% sodium hydrogen carbonate solution (200 ml) and water (50 ml). These extracts were acidified with hydrochloric acid (3 M, 50 ml) and filtered and the residue was dried under vacuum over silica gel to give the title compound (1.105 g, 63% yield based on available bromoacetyl and chloroacetyl compounds) as a pale yellow solid, mp 195-197 0C.
1Ii nmr (400 MHz, dβ-dmso) 8 2.52 (resonance obscured by residual dj-dmso); 2.67 (t, J = 7.0Hz, 2H); 3.96 (s, 2H); 7.19 (s, IH); 8.13 (s, 1 M); 12.32 (br s, 2H).
Example 31
Figure imgf000084_0001
(31)
Preparation of 6-((butylthio)acetyJ)-5-c:h]oro-3-methyl-l,3-beπzothiazoi-2(3fl)-one (31)
(i) 5-Chloro-3-methyϊ-1 ,3-benzothiazol-2(3H)-one 5-Chloro-2-benzothiazolor.e (5.010 g, 27.0 mmol) was dissolved in anhydrous DMF (60 ml) and anhydrous potassium carbonate (11.248 g, 81.4 mmol) and iodomethane (5.05 ml, 81.1 mmol) were added and the mixture stirred at room temperature overnight. The reaction mixture was poured into chloroform (240 ml) and filtered and the filtrate was evaporated to dryness. The resultant residue was dissolved in a mixture of ethyl aceLate (3(X) ml) and water (200 ml) then the phases were separated. The ethyl acetate phase was washed with water (2 x 100 ml) and brine (100 ml), dried over anhydrous magnesium sulfate, and filtered. The filtrate was evaporated to dryness to give the title compound (5.078 g, 94% yield) as a beige powder.
Η nmr (400 MHz, CDC13) δ 3.44 (s, 3H); 7.05 (d, /= 1.6Hz, I H); 7.16 (dd, J= 8.4, 2.0H7., lH); 7.34 (d, /= 8.4H7., I H).
(ii) 6-(Brornoacetyl)-5-ehloro-3-τnethy!- 1 ,3-benzothiazol-2(3tf)-one
Anhydrous DMF (3.0 ml) was added dropwise to aluminium chloride (14.768 g, 111 ωrnol) with .stirring (Caution: exothermic) and the mixture was stirred until an even slurry had formed. 5-Chloro-3-methyl- L ,3-benzothiazυl-2(3/y)-one (2.724 g, 13.6' mmol) was added in portions then the mixture was heated at 70"C under nitrogen. Bromoacctyl bromide (2,0 ml, 23 mmol) was added and heating continued at 70ώC under nitrogen for a further 6h. The mixture was allowed to stand at room temperature under nitrogen overnight then poured onto ice (200 g), stirred for Ih and filtered. The residue was washed with water (2 x 100 ml) and dried at the pump. The residue was then washed with ethyl acetate (2 x 20 ml) and dried at the pump to give the tide compound (2.538 g, 58% yield) as a red-grey solid. ] H nmr analysis showed that the product contained 6-(chloroacctyl)-5-chloro-3-methyl-l,3- ben7.othiazol-2(3//)-one (23 mcil%) and unchanged starting material (6 mol%).
1H nmr (400 MHz7 oVdmso) δ 3.44 (s, 3H); 4.85 (s, 2H); 7.62 (s. IH); 8.24 (s, IH"). Chloroacciyl impurity: δ 3.44 (s, 3H); 5.05 (s, 2H); 7.62 (s, IH); 8.22 (s, IH).
(ϋi) 6-((Butyltl]io)aceiyO-5-chloiO-3-.methyl-l,3-beπ?otliiazol-2(3H)-one (31)
1-Butanethiol (607 mg, 6.73 mmol) was dissolved in anhydrous DMF (20 ml) arid 6- anhydrous potassium carbonate (4.375 g, 31.7 mmol) were added. The mixture was stirred under nitrogen for lOSmin then the reaction mixture was partitioned between ethyl acetate (100 ml) and hydrochloric acid (IM, 80 ml). The aqueous phase was extracted with ethyl acetate (50 ml) and [he ethyl acetate extracts were washed with water (2 x 75 ml) and brine (75 ml), dried over anhydrous magnesium sulfate, and filtered. The filtrate was evaporated to dryness Io give a brown oil that was purified by bulb-to bulb distillation (2350C / O.Sϋmbar) to give the title compound (1.328 g,'65% yield) as a brown oil.
1H nmr (400 MH/., d<,-dmso) δ 0.83 (t, / = 7.2Hz, 3H); 1.29 (sextet, J = 7.5Hz, 2H); 1.47 (quintet, J = 7.4Hz, 2H); 2.47 (resonance obscured by residual d5-dmst)); 3.42 (s, 311); 3.89 (s, 2H); 7.56 (s, 1 H); 8.16 (s, IH).
Example 32
Figure imgf000086_0001
(32)
Preparation of 3-((2-(S-chIoro-3-methyl-2-oxo-2,3-dihydro-l^!-benzothiazol-6-yl)-.2» oxoethyl)thio)propanoic acid (32)
3-Mercaptopropionic acid (515 mg, 4.85 mmol) was dissolved in anhydrous DMF (15 ml) and 6-(bromoacely])-5-chloro-3-methyl-l,3-bcnzothiazol-2(3H)-one (1.551 g, 4.84 mmol) and anhydrous potassium carbonate (3,595 g, 26.0 mmol) were added. The mixture was siirced under nitrogen for 40 mill then the reaction mixture was partitioned between ethyl acetate (150 ml) and hydrochloric acid (IM, 80 ml). The aqueous phase was extracted with ethyl acetate (50 ml) and the combined ethyl acetate extracts were washed with water (2 x 100 ml), and extracted with a 5% sodium hydrogen carbonate solution (200 ml), and water (50 ml). These extracts were acidified with hydrochloric acid (3M, 50 ml) causing a brown oil to precipitate. The mixture was extracted with ethyl acetate (100 ml, 50 ml) and the ethyl acetate extracts were washed with brine (75 ml), dried over anhydrous magnesium sulfate and filtered. The filtrate was evaporated to dryness to give the title compound (1.476 g» 88% yield) as .a cream powder, mp 120-121 0C.
5 1H nmr (400 MHz, dg-dmso) δ 2.53 (resonance obscured by residual ds-dmso); 2.67 (t, J = 7.0Hz, 2H); 3.43 (s, 3H); 3.97 (s, 2H); 7.58 (s, IH); '8.19 (s, IH); 12,31 (br s, U-I).
BIOLOGY EXAMPLES
Example 1. Interaction of com pounds with MIF proteins detected by iSiacore analysis
Methods
J 0 The interaction of compounds with MJF protein was characterized by Surface Plasmon Resonance (SPR) analysis using an S51 (Biacore International AB) automated small molecule biosensor assay system. Recombinant MlH protein was immobilized on a caτboxymethyl dextran biosensor chip using amine coupling chemistry. Compound binding to the immobilized MIF protein was measured at 1.1 concentrations up to 100 uM (in
15 duplicate), with corrections for the DMSO used us a solvent al a final concentration of 5%. The change in SPR output relative to that of a control uπdcrivatized reference spot was recorded over time. The affinity and stoichiometry of interaction was calculated using steady state and/or kinetics evaluation methods with software supplied by tne manufacturer.
Results
20 The results listed in Table 1 summarize the interaction of Compounds 4 , 13, and 19 with immobilized recombinant MIF protein. The compounds bind to MIF with equilibrium dissociation constant (Ko) values in the low micromolar range. The predicted stoichiometry of the compound: MIF turners were determined to be 1 : 1. Table 1. Summary of affinity and kinetic constants for compound binding to immobilized MIF.
Figure imgf000088_0001
Example 2. In vitro assay of MIF antagonism: inhibition of LPS-iπduced production of IL-6 in RAW264.7 macrophages by compounds
MIF is an important factor in the innate immune response to toxins such as the bacterial endotoxin lipopolysaccharide (LPS). Notably, endogenous M (F activity is required for expression of the LPS receptor toll-like receptor-4 ^12I A compound with the ability to inhibit the biological activity of MlF would therefore inhibit the activation of cytokine production by macrophages in response to LPS.
Methods
The RAYV264.7 mouse macrophage cell line was propagated in DMEM/l 0% foetal calf serum (FCS) a. 370C in 5% CO2. 24 hr prior to assay cells were seeded in 96-wcll tissue culture plates. Cells were allowed to adhere for 4 hi prior to transfer to DMEM/0.5% FCS for 18 hr. Cells were then treated with 50 uM compound in DMSO for 30 min prior to stimulation for 4 hr with 100 ng/ml LPS. Cell culture supcm'atants were then collected from each well and assayed for IL-6 levels by ELISA (R&D Systems) according to the manufacturer's Instructions.
Results
Figure 1 shows that Compound 19 treatment induces a dose-dependent inhibition of LPS- induced IL-6 production when RAW264.7 cells are pre-ireated with up to 100 μM concentration of compound and the samples analysed for JL-6 production as described above. The IC50 value for the compound was determined to be 20 uM.
Table 2 shows the % inhibition of IL-6 production induced by 50 uM compound treatment relative to LPS + PMSO control levels (with basal levels of IL-6 in llic absence of LPS subtracted). The compounds induce marked decreases in IL-6 production consistent with antagonism of endogenous MlF. Table 2. Inhibition of LPS-induced IL-6 production in RAW264.7 cells
Figure imgf000090_0001
Example 3. In vitro assay of MIF antagonism: inhibition of interlqukin-l induction of cycloxygenase-2 expression in S 112 human dermal fibroblasts by Compounds
The activity of compounds was studied in a bioassay for MIF-dependent cytokine effects of human Sl 12 dermal fibroblasts. In these cells the induction of the expression of cyclooxygenase-2 (COX-2) protein by interleukin 1 (IL-I) is dependent upon the presence of endogenous MIF CI3). The expression of COX2 proteins is therefore sensitive to depiction of endogenous MIF by neutralizing antibody, gene knockout of targeting with small molecule inhibitors. A compound with the ability to inhibit the biological activity of MIF would therefore inhibit the activation of COX2 expression in response to IL-I.
Methods
Sl 12 human dermal fibroblasts were propagated in RPMI/10% foetal calf serum (FCS). Prior to experimentation, cells were seeded at 105 cells/ml in RPMI/0.1% BSA for 18 hours. Cells were treated with recombinant human IL-I (0.1 ng/ml) and with each compound, at concentrations raingiπg up to 100 μM. A control was treated only with recombinant human IL-I (0.1 ng/ml) and vehicle (DMSO). After 6 hours, cells were collected and intracellular COX-2 protein determined by pcrmeabilisation flow cytometry. Cells permeabilised with 0.1% saponin were sequentially labelled with a mouse anti-human COX-2 monoclonal antibody and with shecp-anti-mouse F(ab)2 fragment labelled with fluorosccin isothioeyanate. Cellular fluorescence was determined using a flow cytometer. At least 5000 events were counted for each reading, each of which was performed hi duplicate, and the results expressed in mean fluorescence intensity (MFI) after subtraction of negative coπtrol- labellcd cell fluorescence.
Results
Figure 2 shows treatment with Compound 2 induces a dose-dependent inhibition of IL-I induced COX-2 expression when S 1 12 cells are treated with up to 100 μM concentration of compound and the samples analysed for COX2 expression as above. The results show significant and dose-dependent reductions in COX2 expression levels consistent with antagonism of M IF activity.
Example 4. In vivo assay of MIF antagonism; Eπdotoxic shock
The activities of compounds were studied in the murine endotoxic shock model. This model has been previously shown to be dependent on M fF (|4). Administration of a compound which inhibits the cytokine activity of MIF would be expected to result in a reduction in serum levels of the pro- inflammatory cytokine TNF. Methods
Endotoxaemia was induced by intra-peritoπeal Injection of C57B1/6J mice with lipopolysaccharidc (LPS) (lmg/kg) in 200 μl saline. Animals were treated with either a saline solution (control) only, or LPS with vehicle or compounds Bl and A3 in vehicle at doses of 10, 1 and 0. t mgΛtg body weighL, administered by intra-peritoncal injection at 24 hours and 1 hour before intra-peritoneal LPS injection. After I hour mice were humanely killed by CO2 inhalation then neck dislocation. Serum was obtained from blood obtained by cardiac puncture prior to death and measured for TNF levels by ELISA according to the manufacturer's instructions.
Results
The results in Figure 3 show that treatment of mice with compounds 15 (Figure 3A) , compounds 2 and 13 (Figure 3B), compound 4 (Figure 3C)1 and compound 19 (Figure 3D) results in a significant dose-dependent suppression of LPS-induced serum TNF levels in the endotoxic shock model described above.
Example 5. Inhibition »f MlF tauttsomerase activity.
MlF protein has the ability In vitro to catalyze the tautisomerizalion of dopachrome ^15\ The tautomerase activity of MIH is unique, as is the structure and sequence of the section of MIF responsible for this phenomenon, suggesting that small molecules binding to or docking in this site would be specific for MIF. The relevance of this enzymatic activity to the development of inhibitors of the cytokine and biological activity of MIF is that demonstration of inhibition of taulisonαerαse activity is a demonstration that a given compound has a direct physical interaction with MΪF.
Methods
.Recombinant human MIF protein was pre-incubated with compounds as indicated prior to the addition of J, -dopachrome substrate. Tautomerase activity was determined by measurement of the decrease in absorbance at 475 nni after 2 mini The maximum tautisomerase activity detected was recorded as 100%, «uid the inhibition of this activity at cither 50 iuM or 100 tnM concentration of compounds determined.
Results
Many compounds were determined to bind to MBF via demonstration of the ability to inhibit the tautisomerase activity of MIF, as shown in Table 3. Values shown arc the mean ± standard deviation of 2-4 experiments.
Table 3: Inhibition of the tautisomerase activity oi" MIF by selected examples
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Example 6
Delayed-type hypersensitivity reactions, which are itiitatcd by T lymphocyte responses to recall antigens and mediated by many cell types including macrophages, are known to be dependent on the cytokine or biological activity of MIF (16' I7). For example, an anti-MIF monoclonal antibody suppresses delaycd-typc hypersensitivity reactions reactions in vivo to methylated bovine scrum albumin (mBSA) injected inio the skin of animals preimmunised with mBSΛ 06K A compound inhibiting Lhc cytokine or biological function of MIF might be expected to inhibit delayed -type hypersensitivity reactions in vivυ.
Methods
Mice were immunised on day 0 with 200 μg of methylated BSA (niBSA;.Sigma Chemical Co., Castle Hill, Australia) emulsified in 0.2 ml of Freuπd's complete adjuvant (CFA; Sigma) injected subculaneously in the flank skin. At day 7, mice were given 100 μg tnBSA in 0.1 ml CFA by intradermal injection at the base of the tail. Mice were challenged on day 27 following first immunisation by a single intradermal (ID) injection of 50 μg mBSΛ/20 μl saline in the right footpad, with 20 μl saline injected in the left footpad serving as control (Santos, 2001). Mice were killed 24 h later and footpad swell ing quantified using micro calipers (Mitutoyo, Kawasaki-shi, Japan). DTH measurements were performed by an observer blinded to mouse genotype. Results were expressed as the difference in footpad swelling between πiBSA and saline-injected footpads, and expressed as change in footpad thickness (mm). Mice were treated with compound 13 at 5 and 15 mg/kg/24h by EP injection, twice daily for 7 days prior to antigen challenge with rnBSA in the footpad. Treatment with compound 13 continued for a further 24h and changes in footpad thickness relative to control paws were measured at that time. As shown in Figure 4, compound 13 induced a significant inhibition of DTH reactions.
Example 7
MIF is implicated in lhe recruitment of leukocytes to sites of inflammaύon, via studies which show that MIF-deficient mice exhibit reduced interactions between leukocytes and vascular endothelium in vzWls>. More recently, it has been demonstrated that the administration of MIF in vivo induces the recruitment of macrophages to tissue (l9\ a process which first requires the induction of adherence oϊ circulating leukocytes to the vascular endothelial cells. As will be known to those skilled in the art, the adherence of leukocytes to the endothelium in vivo can be sLudied using the technique of intravital microscopy (l8-19). As MIF induces leukocyte adherence to vascular endothelium as measured using intravital microscopy, a compound inhibiting the Cytokine or biological activity of MIF might be expected to inhibit the effects of MIF observable using intravital microscopy.
Methods
Mice were anesthetised with kelamine/xylazinc, and the crcmastcr muscle was exteriorized onto an optically-clear viewing pedestal. The cremasteric microcirculation was visualized using an intravital microscope (Axioplan 2 Imaging; Carl Zeiss, Australia) with a 2OX objective lens (LD Achroplaii 20X/0.40 NA, Carl Zeiss, Australia) and a 1OX eyepiece. Three-five postcapillary venules (25-40 μm in diameter) were examined for each experiment. Images were visualized using a video camera and recorded on video-tape for subsequent playback analysis. Recombinant human MTF (1 mg) was injected inlrascrolally in 150 μL saline, prior to intravital microscopy 4 hours later. Leukocyte-endotheUal cell adhesion, was assessed as described by Gregory et al (l9\ Compound 13 at a dose of 30 mg/kg or vehicle were administered by intraperitoneal injection 10 minutes prior to intrascroial injection of MIF.
Results
As shown in Figure 5, MIF induced leukocyte adhesion markedly above baseline leukocyte adhesion observed without MIF injection (dotted line). MlF-induced leukocyte adhesion was reduced approximately 50 % by compound 13 administration. These results arc consistent with inhibition by compound 13 of In vivo effects or esogenously administered MTF.
Determination of lower limits of solubility of compounds
An important physicochemical characteristic of pharmaceutical compounds is that the aqueous solubility of the compound is sufficiently high to allow dosing of humans with a pharmacologically active dose. Compounds with only limited aqueous solubility may be less suitable for development as a human therapeutic.
Methods
Lower limits of aqueous compound solubility were determined in a nepholometer in phosphate-buffered saline containing ϋ.005% (v/v) P20 and a. final concentration of 5% DMSO. Briefly, compounds were initially dissolved in DMSO as a 10 mM stock solution and diluted lo 1 mM. and 0.5mM working solutions with neat DMSp. The compounds were then titrated in DMSO and a constant volume of DMSO stock added to filtered PBS/P20 solution so that the final DMSO concentration was 5%. The solubility was then determined in clear, flat-bottom 96- well plates using the nephclometer and reported as the concentration range at which the compound begins to precipitate from solution. Results
The results in Table 4 show that these compounds have excellent solubilities which would support dosing in humans in the iiM drug range.
Table 4. Solubility assessment of compounds using nephelometry
Figure imgf000098_0001
Throughout tills specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other clement, integer or step, or group of elements, integers or steps.
AU publications mentioned in this specification are herein incorporated by reference. Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context tor the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed in Australia or elsewhere before the priority date of each claim of this application.
It will be appreciated by persons skilled in. the art that numerous variations and/or departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
REFERENCES
L David, J, Proc. Natl. Acad. Sci., USA, (1966), 56, 72-77.
2. Leiser, W. Y., el al., Proc. Natl. Acad. Sci., USA, (1989), 86, 7522-7526.
3. Leech M, Metz CN, Smith M, Weedon H, Holdsworth SR, Bueala R, et al. Macrophage migration inhibitory factor (MIF) in rheumatoid arthritis: Evidence for pro-inflammatory function and regulation by glucocorticoids. Arthritis & Rheumatism 1999; 42: 1601-1608.
4. Morand EF1 Bueala R, Leech M. Macrophage migration inhibitory factor (MfF): An emerging therapeutic target in rheumatoid arthritis. Arthritis & Rheumatism 2003; 48:291-299.
5. Calandra T, Bernhagen J, Met/ CN, Spiegel LA, Bachcr M, Donnelly T, et al. MIF as a glucocortieoid-induced modulator of cytokine production. Nature 1995; 377:68-71.
6. Donnelly SC, Haslell C, Reid PT, Grant fS, Wallace WAH, Metz CN. I. Regulatory role for macrophage migration inhibitory factor in acute respiratory distress syndrome. Nature Medicine 1997; 3:320-323.
7. Bacher M, Metz CN, Calalidra T, Mayer K, Chcsney J, Lohoff M, eL al. An essential regulatory role for macrophage migration inhibitory factor in T-cell activation. Proceedings of the National Academy of Sciences USA 1996; 3:7849-7854. 8. Santos LL, Hall P, Metz CN, Bueala R, Morand HF. Role of macrophage migration inhibitory factor (MIF) in murine antigen-induced arthritis: Interaction with glucocorticoids. Clin.Exp.Immunol. 2001; 123:309-314.
9. Leech M, vSantos LL, Met? C, Holdsworth SR, Bueala R, Morand EF. Control of macrophage migration inhibitory factor (MTJ7) by endogenous glucocorticoids in rat adjuvant arthritis. Arthritis & Rheumatism 2000; 43:827-833.
10. Bueala R. MfF rediscovered: cytokine, pituitary hormone, and glucocorticoid-lnduced regulator of the immune response: FASEB.J. 1996; 10:1607-1613. 11. Sabroe lv Pease JE, Williams TJ , Asthma and MIF: innately Th 1 and Tϊϊ2. Clin Exp Allergy 2000; 30(9): 1194-6.
12. Roger, T., J. David, M. P. Glauser , and T. Calandra. 2001. MIF regulates innate immune responses through modulation of Toll-like receptor 4. Nature 414:920. 13. Sampcy. A. V., P. H. Hall, R. A. Mitchell, C. N. Metz, and E. F. Moraad. 2001.
Regulation of synoviocyte phospholipase A2 and cyclooxygenasc 2 by macrophage migration inhibitory factor. Arthritis Rheum 44:1273.
14. Bcmhagen, J., T. Calandra, R. A, Mitchell, S. B. Martin, K. J. Traccy. W. Voelter, K, R. Manogue, A. Ccrami; and R. Bucala. 1993. MlF is a pituitary-derived cytokine . that potentiates lethal endotoxaemia. Nature 365: 756.
15. Rosengrcn, E.v R. Bucala, P. Aman, L. Jacobsson, O. Odh, C. N. Metz, and H. Rorsman. 1996. The imnαunoregulatory mediator macrophage migration inhibitory factor (MIF) catalyzes a tautomerization reaction. MoI Med 2:143-149.
16. Santos, L. L., P. Hall, C. N. Metz, R. Bucala, and E, F. Morand. 2001. Role of . macrophage migration inhibitory factor (MIF) in murine antigen-induced arthritis:
Interaction with glucocorticoids, Clin Exp Immunol 123:309-314.
17. Bernhagen, J., M. Baciier, T. Calandra. C. N. Metz, S. B. Doty, T. Donnelly, and R. Bucala. 1996. An essential role for macrophage migration inhibitory factor in the tuberculin del ayed-type hypersensitivity reaction. J Exp Med 183:277-282. 18. Gregory, J. L., M. T. Leech, J. R. David, Y. H. Yang, A. Dacumos, and M. J . Hickey. 2004. Reduced lcukocyte-endothelial cell interactions in the inflamed microcirculation of macrophage migration inhibitory factor-deficient mice. Arthritis Rheum 50:3023-3034.
19. Gregory, J. L., E. F. Morand, S. J. McKeown, J. Λ. Ralph, P. Hall, Y. H. Yang, S. R. McCoIl, and M. J. Hickey. 2006. Macrophage Migration Inhibitory Factor Induces
Macrophage Recruitment via CC Cbemokinc Ligaπd 2. J Immunol 177:8072-8079. 20. Λcberli D, Yang YH, M.ansell A7 Santos L, Leech M, Morand EF; Macrophage migration inhibitory factor modulates glucocorticoid sensitivity Ln macrophages via effects on MAP kinase phosphatase- 1 and p3S MAP kinase. FEBS Lett 2006, 580:974-981. 21. Roger T, Chanson AL, Knaup-Rcymond M, Calandra T: Macrophage migration inhibitory factor promotes innate immune responses by suppressing glucocorticoid- induccd expression of mitogen-activated protein kinase phosphatase-! . Eur J Immunol 2005, 35:3405-3413.
22. Gregory J L, Morand EF, McKeown SJ, Ralph JA, HaII P, Yang YH7 McCoIl SR, Hickey MJ: Macrophage Migration Inhibitory Factor Induces Macrophage
Recruitment via CC Chcniokine Ligand 2. J Immunol 2006, 177:8072-8079.
23. Gregory JL, Leech MT, David JR7 Yang YJ I, Dacumos A, Hickey MJ: Reduced leukocyte-endothelial cell interactions in the inflamed microcirculation of macrophage migration inhibitory factor-deficient mice. Arthritis Rheum 2004, 50:3023-3034.
24. Morand EF, Leech M, Berπliagen J : MIF: a new cytokine link between rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov 2006, 5:399-410.

Claims

L A method of treating, diagnosing or preventing autoimmune diseases, tumours, or chronic or acute inflammatory diseases comprising administering a treatment, prevention or diagnostic effective amount of a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof wherein:
Figure imgf000103_0001
X is selected from- -O-, -S-, -C(R5)(Rs-)- and -N(Rn)-;
Y is selected from - -N(R7)-, -O-, -S-, and -C(R7)2-;
Z is selected from >C=O, >C=S, >C=NR6, >S=O and >S(O)a;
R1 is selected from hydrogen, C|-Q,alkyl, (CR5R5XiOR7, C(R5R5OnSR7, (CR5R5OaN(Ro)2 and (CR5R50πhulo;
R3 is selected from hydrogen, Ci-Q,alkyl, (CR1(JR16)PNR14R15, (CR1nR16-OpPR17, (CR16RIeOpSR17, (CR1eR1eOphalo,
Figure imgf000103_0002
(CR16R10OnC(O)R28,
Figure imgf000103_0003
(CR1 nR16OnS(O)R175 (CR1SR16OnS(O)2R17, (CR16R10OnS(O)3R17, and (CR1fiRw0pC(R1«)3; R4 is selected from hydrogen, halogen, Ci-Cjalkyl, Cr-Qjalkcnyl, CrCjalkytiyl and
Figure imgf000104_0001
each R5 and Rs> is independently selected from hydrogen, Ci-Csalkyl, halo, OR7, SR7 and N(Re)2;
each Rβ is independently selected from hydrogen, Ci-Cjalkyl and OR7;
each R7 is independently selected from hydrogen and Ci-C3alkyl; •
each R12 and R12' is independently selected from hydrogen, Ci-Csalkyl, C2-Cftalkcnyt, CVQsalkynyl, OR24, SR24, halo, N(R24)2, CO2R21, CN, NO2, aryl and hctcrocyclyl;
each R14 and Ru is independently selected from hydrogen, Ci-C3alkyl, OR17, SR17, and N(R17).;
each R10 and R^ is independently selected from hydrogen, Cι-QjaLkyl, halo, OR17, SR17 and N(RI7)2;
each R17 is independently selected from hydrogen and CrC3alkyl;
each R1H is independently selected from hydrogen and halo;
R22 is selected from Cι-C6alkyl, NH2, N! r(C,-CήalJ-:yl), N(Cι-Cftalkyl)2, OR29 or SR29;
each. R24 is selected from H and Ci-Cgalkyl;
R28 is selected from hydrogen, Ci-C<>alkyl, OR29, SRM or N(R^)2;
each R29 is independently selected from hydrogen and Ci-Caalkyl;
Q is selected from O, S, NR40, S(O)11 where u is an integer from 1 to 2;
R40 is selected from H, OH, and
Figure imgf000104_0002
.
each R41 and ΕUv is independently selected from H, OfT, halo, NH2, cyano, and NO2; R42 is independently selected from H, 0R«, COOR4:!, CON(R43R^'), 0(CO)R-U, aryl, and heterocyclyl;
each R4J and R<t.τ is independently selected from M,
Figure imgf000105_0001
benzyl, and aryl;
n = O or ail integer 10 3
m is O or an integer from 1 to 20;
p is O or an integer from 1 to 6;
t is an integer from 1 tol 0
v is 0 or an integer from 1 to 10.
2. A method according to claim 1, wherein the autoimmune disease, tumour, or chronic or acute inflammatory disease is selected from the group consisting of:
rheumatic diseases; spondyloarthropathies; crystal arthropathies; Lyme disease; polymyalgia rheumatica; connective tissue diseases; vasculitldes; inflammatory conditions; sarcoidosis; vascular diseases; vascular occlusive disease; vascular stent restenosis; ocular diseases; auioimmuue diseases; pulmonary diseases; cancers; renal diseases; disorders of the hypotJhalamie-pituitary-adrcaial axis; nervous system disorders; diseases characterised by modified angiogenesis; endometrial function; complications of infective disorders; transplant rejection, graft- versus-host disease; allergic diseases; bone diseases; .skin diseases; diabetes mellilus and its complications; pain, testicular dysfunctions and wound healing; gastrointestinal diseases; peptic ulceration; gastritis; oesophagitis; and liver disease.
3. A method according 10 claim 1 or claim 2, wherein MlF cytokine or biological activity is implicated in the disease or condition.
4. A method according to any one of claims 1 to 3, wherein the disease or condition is selected from the group consisting of rheumatoid arthritis, systemic lupus uveitis, diabetes tneliitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and chronic obstructive pulmonary disease.
5. A method according to uny one of claims 1 to 4, wherein Q is S.
6. A method according Io any one of claims 1 to 5, wherein R«ι is C(Rn R4 y)vRi2 and R42 is COOR43.
7. A method according to claim 6, wherein R4^ is hydrogen or Ci-Cgalkyl,
8. A method according to claim 6 or claim 7, wherein R43 is methyl.
9. A method according to any one of claims 1 to 4, wherein the compound of formula (I) is selected from the group consisting of:
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
10. A method according to claim 9, wherein the compound of formula ([) is selected from the group consisting of:
Figure imgf000108_0002
.1 L A compound selected from the group consisting of:
Figure imgf000108_0003
Figure imgf000109_0001
Figure imgf000110_0001
12. A compound of Formula (E) or a pharmaceutically acceptable salt or prodrug thereof wherein:
Figure imgf000110_0002
II
X is selected from - -O-, -S-, -C(R5)(R5')- and -N(R6)-;
Y is selected from - -N(R7)-, -O-, and -S-;
Z is selecLed from >C=O, >C=S, and >C=NR&; R, is selected from hydrogen, C)-C3alkyl, (CR5R5OnOR7, C(R5RsOuSR?, (CR5RsOoN(Rs)2 and (CR5Ry)JIaIo;
R3 is selected from hydrogen, Ci-Cfialkyl, (CR, ΛR, ^
Figure imgf000111_0001
)11N R ,4R15, (CR)6R16OpSR17, {CRJ6Rt6')phalo, (CR16R16OpNO2, (CR16R^)nC(O)R2S,
Figure imgf000111_0002
(CR1 6R16OnS(O)3R17, and (CR16R16OpC(R1S)3;
R4 is selected from hydrogen, halogen, C]-C3alkyl, Ca-C^alkeπyK Cs-Cjalkynyl and
Figure imgf000111_0003
each R5 and Rs- is independently selected from hydrogen, Cf-Ojalkyl, halo, OR7, SR7 and N(Rβ)2;
each R(, is independently selected from hydrogen, Ci-Caalkyl and OR7;
each R7 is independently selected from hydrogen and Ci-G)alkyl;
each R 12 and Riz- is iαdependently selected from hydrogen, Ci-Cgalkyl, Ca-CήSlkenyl, Ca-Cealkyiiyl, OR24, SRM, halo, N(R2^)2, CO2R24, CN, NO2, aryl and heterocyclyl;
each R]4 and R15 is independently selected from hydrogen, Cj-Cjalkyi, ORf7, SR17, and N(Rn)2;
each R1 fj and Rw is independently selected from hydrogen, C|-C3alkyl, halo, OR17, SR17 and N(Rn)2;
each Rn is independently .selected from hydrogen and
Figure imgf000111_0004
each R is is independently selected from hydrogen and halo;
R22 is selected from Ct-Cfialkyl, NH2, NH(d-Cftalkyl),
Figure imgf000111_0005
OR29 or SR29;
each R24 is selected from H and Ci-Qalkyl; R2S is selected from hydrogen, Ci-Cgalkyl, OR29, SR20 or N(R2?).;
each B-29 is independently selected from hydrogen and C|-C3alkyi;
Q is selected from O , S , S(O)1, where u is an integer from 1 102;
R40 is selected from H, OH, and C(R4I R410vR42;
each R4i and R41' is independently selected from H, OH, halo, NIb1 CN and NO2:
R42 is selected from. H, OR43, COOR43, CON(R4SR4T), 0(CO)R43, N(R4SR**), aryl, and hcterocyclyl;
each R4J and R43' is independently selected from H, C1^ alfcyl, and benzyl;
n is O or 1 to 3;
m is O or an integer from 1 Lo 8;
p is O or an integer from 1 to 6;
t is an integer from 1 to 10;
v is O or an integer from 1 to 10
provided that the compound is not
Figure imgf000112_0001
13. A compound according to claim 12, wherein Q is S.
14. A compound according Lυ claim 12 or claim 13, wherein R^i is C(R41R4IOvR^ and R42 is COORo.
15. A compound according to claim 14, wherein R4J is hydrogen or Ci-Qjalkyl,
16. A compound according to claim 14 or claim 15, wherein R^ is methyl.
17. Λ compound of Formula III or a pharmaceutically acceptable salt or prodrug thereof wherein:
Figure imgf000113_0001
UI
X is selected from - -O-, -S-, -C(R5)(Ry)- and -N(R6)-; .
Y is selected from - -N(R?), -O-, and -S-;
Z is selected from >C=O, >C=S, and >C=NRe;
R, is selected from hydrogen, Ci-C3alkyi, (CR5R-T)11OR7, C(R5R5OnSR7, (CR5R5OnN(R5)Z and (CR1 R5OJIaIo;
R3 is selected from hydrogen, C-Qalkyl, (CRI6RJ0OPNR]4RJS, (CR10R1βOpORπ, (CR16R16OpSR17, (CR1AR16.)phιιlo, (CR16R16O1^O2, (CR,6R16-λιC(O)R,
Figure imgf000113_0002
(CR16R16OS(O)R17, (CR16RIn)nS(O)2Rn, (CR16R1^)nS(O)3R17, and (CR,c,R]60pC(R,K).i; R4 is selected From hydrogen, halogen,
Figure imgf000114_0001
C2-C3aDtcnyl, C2-Qjalkynyl and
Figure imgf000114_0002
each R5 and R5' is independently selected from hydrogen, Ci-C3alkyl, halo, OR7, SR7 and N(Re)2;
each Rg is independently selected from hydrogen, Ci-Cjalkyl and OR7;
each R7 is independently selected from hydrogen and Cι-C3alkyl;
each R12 and R12 1 is independently selected from hydrogen, Ci-Csalkyl, C2-Q,alkeny(, C2"C6alfcynyl, OR24, SR24, halo, N(R24)., CO2R24, CN, NO2, aryl and heterocyclyl;
cachR14 and R15 arc independently selected from hydrogen, Ci-C^alkyl, OR17, SR17, and N(R, 7)2;
each R10 and R1β' is independently selected from hydrogen, d-C3alkyl, halo, OR17, SR17 and N(Rn)2;
each Rn is independently selected from hydrogen and Ci-Cjalkyl;
each R1g is independently selected from hydrogen and halo;
R22 is selected from d-Cgalkyl, NH2, NH(C,-Cfialkyl), N(C,-Q,alkyl)2, OR29 or SR29;
each R24 is selected from H and Ci-Qalkyi;
R2x is selected from hydrogen, Ci-Cgalkyl, OR29, SR29 or N(R29)2;
each R29 is independently .selected from hydrogen and Cι-C3alkyl;
R44 is selected from OH, C(R45R45OvR4C;
each R45 and R45' is independently selected from H, OH, halo, NH2, CN, NO2;
each R40 is selected from COOR47, CON(R47R47O,. O(CO)RΛ7, N(R47R47O; each B47 and R47' is independently selected from H, Cj.g alkyl, benzyl;
wherein when v is greater than 1 , R4^ can be OR47;
wherein when v is greater than 2 , R4^ can be H;
Λ is O OT 1 lo 3;
in is O or an integer from I to 8;
p is O or an integer from 1 to 6;
t is an integer from 1 to 10;
v is 0 or an integer from 1 to 10
provided that, flic compound is not
Figure imgf000115_0001
18. A use of a compound of Formula (1) us defined in claim 1 , or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for treating, diagnosing or preventing autoimmune disease, tumour, or chronic or acute inflammatory disease selected from the group consisting of:
rheumatic diseases; spondyloarthropathies; crystal arthropathies; Lyme disease; polymyalgia mcumatiea; connective tissue diseases; vasculm'des; inflammatory conditions; sarcoidosis; vascular diseases; vascular occlusive disease; vascular stent restenosis; ocular diseases; autoimmune diseases; pulmonary diseases; cancers; renal diseases; disorders of the hypothalamic-pituitary-adrenal axis; nervous system disorders; diseases characterised by modified angiogenesis; endometrial function; complications of infective disorders; transplant rejection, graft-versus-host disease; allergic diseases; bone diseases; skin diseases; diabetes mcllitus and its complications; paiα, testicular dysfunctions and wound healing; gastrointestinal diseases; pcpLic ulceration; gastritis; oesophagitis; and liver disease.
19. A use according to claim 18, wherein MlF cytokine or biological activity is implicated in the disease or condition.
20. A use according to claim 18, wherein the disease or condition is selected from the group consisting of rheumatoid arthritis, systemic lupus ery thematosυs, ulcerative colitis, Crohn's disease, midliple sclerosis, psoriasis, uveitis, diabetes mellitus, glomerulonephritis, atherosclerotic vascular disease and infarction, asthma and . chronic obstructive pulmonary disease.
21. A pharmaceutical composition comprising a compound according to any one of claims 1 1 to 17 and a pharmaceutically acceptable carrier, diluent or excipient.
22. A method of inhibiting cytokine or biological activity of MIF comprising contacting MIF with a cytokine or biological inhibiting amount of a compound of Formula (I) as defined in claim 1 < or a pharmaceutically acceptable salt or prodrug thereof.
.
23. A method of treating, preventing or diagnosing a disease or condition wherein MIF cytokine or biological activity is implicated comprising the administration of a treatment, prevention or diagnostic effective amount of a compound of Formula (T) as defined in claim 1 , or a pharmaceutically acceptable salt or prodrug thereof to a subject in need thereof.
24. A method of treating or preventing a disease or condition wherein MIF cytokine or biological activity is implicated comprising:
administering to a mammal a compound of Formula (I) as defined in claim 1, or a pharmaceutically acceptable salt or prodrug thereof and a second therapeutic agent,
25. A method of prophylaxis or treatment of a disea.se or condition for which treatment with a glucocorticoid is indicated, said method comprising: administering to a mammal a glucocorticoid and a compound of Formula (I) as defined in claim 1 > or a pharmaceutically acceptable salt or prodrug thereof.
26. A method of treating steroid-resistant diseases comprising:
administering to a mammal a glucocorticoid and a cumpound of Formula (I) as defined in claim 1 , or a pharmaceutically acceptable salt or prodrug thereof.
27, A method of enhancing the effect of a glucocorticoid in mammals comprising administering a compound of Formula (I) as defined in claim 1, or a pharmaceutically acceptable salt or prodrug thereof siinulraneously, separately or sequentially with said glucocorticoid.
28. A pharmaceutical composition comprising a glucocorticoid and a compound of
Formula (I) as defiucd in claim 1, or a pharmaceutically acceptable salt or prodrug thereof.
29. A use of a glucocorticoid in the manufacture of a medicament for administration with a compound of Formula (I) as defined in claim i, or a pharmaceutically acceptable salt or prodrug thereof for the treatment or prophylaxis of a disease or condition, for which treatment with a glucocorticoid is indicated.
30. A use of a compound of Formula (I) as defined in claim 1, or a pharmaceutically acceptable salt or prodrug (hereof in the manufacture of a medicament for administration with a glucocorticoid for the treatment or prophylaxis of a disease or condition for which treatment, of a glucocorticoid is indicated.
31. A use of a glucocorticoid and a compound of Formula (I) as defined in claim 1, or a pharmaceutically acceptable salt or prodrug thereof in the manufacture of a medicament for the treatment or prophylaxis of a disease or condition for which treatment with a glucocorticoid is indicated.
32. An implantable device comprising: (i) a reservoir containing at least one compound of Formula (T) as defined in claim I , or a pharmaceutically acceptable salt or prodrug thereof; and
(ii) means to release or elυte the inhibitor from the reservoir.
33. A method according to claim 32, wherein the implantable device is a stent.
34. A method for inhibiting the cytokine or biological activity of MIF in a subject comprising the step of implanting an implantable device according to claim 32 or claim 33, in n subject.
PCT/AU2006/001965 2005-12-21 2006-12-21 Mif inhibitors WO2007070961A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US12/158,563 US20090130165A1 (en) 2005-12-21 2006-12-21 MIF Inhibitors
EP06840387A EP1968576A4 (en) 2005-12-21 2006-12-21 Mif inhibitors
AU2006326850A AU2006326850A1 (en) 2005-12-21 2006-12-21 MIF inhibitors
CA002634212A CA2634212A1 (en) 2005-12-21 2006-12-21 Mif inhibitors
JP2008546033A JP2009521415A (en) 2005-12-21 2006-12-21 MIF inhibitor
IL192331A IL192331A0 (en) 2005-12-21 2008-06-19 Mif inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US75235405P 2005-12-21 2005-12-21
US60/752,354 2005-12-21

Publications (1)

Publication Number Publication Date
WO2007070961A1 true WO2007070961A1 (en) 2007-06-28

Family

ID=38188170

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2006/001965 WO2007070961A1 (en) 2005-12-21 2006-12-21 Mif inhibitors

Country Status (10)

Country Link
US (1) US20090130165A1 (en)
EP (1) EP1968576A4 (en)
JP (1) JP2009521415A (en)
KR (1) KR20080090435A (en)
CN (1) CN101410107A (en)
AU (1) AU2006326850A1 (en)
CA (1) CA2634212A1 (en)
IL (1) IL192331A0 (en)
WO (1) WO2007070961A1 (en)
ZA (1) ZA200805386B (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010021693A2 (en) 2008-08-18 2010-02-25 Yale University Mif modulators
EP2198879A1 (en) 2008-12-11 2010-06-23 Institut Curie CD74 modulator agent for regulating dendritic cell migration and device for studying the motility capacity of a cell
WO2013163758A1 (en) 2012-05-01 2013-11-07 Boyd Shelley Romayne Methods for treating and diagnosing blinding eye diseases
EP2944310A1 (en) 2014-05-16 2015-11-18 Mifcare MIF inhibitors for the acute or chronic treatment of pulmonary hypertension
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
US9617212B2 (en) 2013-12-17 2017-04-11 Controlled Chemicals, Inc. Isoindolin-1-ones as macrophage migration inhibitory factor (MIF) inhibitors
US9643922B2 (en) 2008-08-18 2017-05-09 Yale University MIF modulators
US11660266B2 (en) 2018-04-11 2023-05-30 Ohio State Innovation Foundation Methods and compositions for sustained release microparticles for ocular drug delivery

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7709514B2 (en) * 2002-06-07 2010-05-04 Cortical Pty Ltd Therapeutic molecules and methods-1

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1982001186A1 (en) * 1980-09-29 1982-04-15 Busch N Novel benzoxazolinones,substituted in the 6-position by an aminoalcohol chain or amino-ketone chain,their preparation and their use in pharmaceutical preparations
JPS60130573A (en) * 1983-12-15 1985-07-12 Otsuka Pharmaceut Co Ltd Benzothiazole derivative
JPH04342569A (en) * 1991-05-17 1992-11-30 Yoshitomi Pharmaceut Ind Ltd Quinolinone compound
WO2003057254A1 (en) * 2001-12-28 2003-07-17 Takeda Chemical Industries, Ltd. Preventives/remedies for urinary disturbance
WO2005000354A1 (en) * 2003-06-30 2005-01-06 Takeda Pharmaceutical Company Limited Preventive/remedy for urinary disturbance
WO2005058304A1 (en) * 2003-12-17 2005-06-30 Cortical Pty Ltd Implantable device containing inhibitor of macrophage migration inhibitory factor
WO2006070934A1 (en) * 2004-12-28 2006-07-06 Takeda Pharmaceutical Company Limited Crystal of bicyclic fused heterocyclic derivative, process for production thereof, and use thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7709514B2 (en) * 2002-06-07 2010-05-04 Cortical Pty Ltd Therapeutic molecules and methods-1
US7216778B2 (en) * 2004-11-29 2007-05-15 Todd Kaeb Self-leveling drip catcher for fluid containers

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1982001186A1 (en) * 1980-09-29 1982-04-15 Busch N Novel benzoxazolinones,substituted in the 6-position by an aminoalcohol chain or amino-ketone chain,their preparation and their use in pharmaceutical preparations
JPS60130573A (en) * 1983-12-15 1985-07-12 Otsuka Pharmaceut Co Ltd Benzothiazole derivative
JPH04342569A (en) * 1991-05-17 1992-11-30 Yoshitomi Pharmaceut Ind Ltd Quinolinone compound
WO2003057254A1 (en) * 2001-12-28 2003-07-17 Takeda Chemical Industries, Ltd. Preventives/remedies for urinary disturbance
WO2005000354A1 (en) * 2003-06-30 2005-01-06 Takeda Pharmaceutical Company Limited Preventive/remedy for urinary disturbance
WO2005058304A1 (en) * 2003-12-17 2005-06-30 Cortical Pty Ltd Implantable device containing inhibitor of macrophage migration inhibitory factor
WO2006070934A1 (en) * 2004-12-28 2006-07-06 Takeda Pharmaceutical Company Limited Crystal of bicyclic fused heterocyclic derivative, process for production thereof, and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP1968576A4 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2326631A2 (en) * 2008-08-18 2011-06-01 Yale University Mif modulators
JP2012500260A (en) * 2008-08-18 2012-01-05 イェール・ユニヴァーシティー MIF modulator
EP2326631A4 (en) * 2008-08-18 2012-03-21 Univ Yale Mif modulators
US11584717B2 (en) 2008-08-18 2023-02-21 Yale University MIF modulators
US10202343B2 (en) 2008-08-18 2019-02-12 Yale University MIF modulators
US9540322B2 (en) 2008-08-18 2017-01-10 Yale University MIF modulators
WO2010021693A2 (en) 2008-08-18 2010-02-25 Yale University Mif modulators
US9643922B2 (en) 2008-08-18 2017-05-09 Yale University MIF modulators
EP2198879A1 (en) 2008-12-11 2010-06-23 Institut Curie CD74 modulator agent for regulating dendritic cell migration and device for studying the motility capacity of a cell
EP3338776A1 (en) 2012-05-01 2018-06-27 Translatum Medicus Inc. Methods for treating and diagnosing blinding eye diseases
WO2013163758A1 (en) 2012-05-01 2013-11-07 Boyd Shelley Romayne Methods for treating and diagnosing blinding eye diseases
US9617212B2 (en) 2013-12-17 2017-04-11 Controlled Chemicals, Inc. Isoindolin-1-ones as macrophage migration inhibitory factor (MIF) inhibitors
WO2015173433A1 (en) 2014-05-16 2015-11-19 Mifcare Mif inhibitors for the acute or chronic treatment of pulmonary hypertension
EP2944310A1 (en) 2014-05-16 2015-11-18 Mifcare MIF inhibitors for the acute or chronic treatment of pulmonary hypertension
US11660266B2 (en) 2018-04-11 2023-05-30 Ohio State Innovation Foundation Methods and compositions for sustained release microparticles for ocular drug delivery

Also Published As

Publication number Publication date
US20090130165A1 (en) 2009-05-21
KR20080090435A (en) 2008-10-08
EP1968576A1 (en) 2008-09-17
AU2006326850A1 (en) 2007-06-28
IL192331A0 (en) 2008-12-29
ZA200805386B (en) 2009-04-29
EP1968576A4 (en) 2010-07-21
JP2009521415A (en) 2009-06-04
CN101410107A (en) 2009-04-15
CA2634212A1 (en) 2007-06-28

Similar Documents

Publication Publication Date Title
EP1968576A1 (en) Mif inhibitors
KR101972719B1 (en) Androgen receptor modulators and uses thereof
KR100926399B1 (en) Diarylamine-containing compounds and compositions, and their use as modulators of steroid hormone nuclear receptors
JP6317320B2 (en) Salt of epidermal growth factor receptor kinase inhibitor
AU2016229136A1 (en) DNA alkylating agents
CS202515B2 (en) Method of producing novel 4-hydroxy-2h-1,2-benzothiazine-3-carboxamide-1,1-dioxides
CA2956465A1 (en) Cyclohexyl-ethyl substituted diaza- and triaza-tricyclic compounds as indole-amine-2,3-dioxygenase (ido) antagonists for the treatment of cancer
CN104619327B (en) Prodrug as the amido quinazoline of kinase inhibitor
EA008865B1 (en) 2-imino-4-oxothiazoline derivatives
CN1240432A (en) 1-phenylpyrazole compounds and medicinal application thereof
JP5227179B2 (en) 3,6-dihydro-2-oxo-6H-1,3,4-thiadiazine derivative
DE102005055354A1 (en) Substituted 5-phenyl-3,6-dihydro-2-oxo-6H- [1,3,4] thiadiazines
WO2019161803A1 (en) Peptidylarginine deiminase inhibitor and use thereof
US10626115B2 (en) Fused pyrimidinone and triazinone derivatives containing bridged nitrogen, their process of preparation and their therapeutic uses as antifungal and/or antiparasitic agents
JPH11513993A (en) Proton pump inhibitor
US6858621B2 (en) 2-(quinolonyl)-fused heterocycles as androgen receptor modulators
ES2294721T3 (en) TIENOPIRICARBOXAMIDAS AND ITS MEDICAL USE.
JPS62503030A (en) Triazolylquinoline derivative
AU2019395282A1 (en) Organophosphorus-substituted compounds as c-MET inhibitors and therapeutic uses thereof
US20230159468A1 (en) Novel forms of pracinostat dihydrochloride
JPH05112559A (en) 4-amino-5-pyrimidinecarboxylic acid derivative
Palled et al. Synthesis of 1, 2, 3, 4-[1-N-Methyl Benzimidazole] 3-Phenyl Substituted Thiazolidine, 4-One and Carbohydrazide Derivatives for Antimicrobial Activity.
JPH02275870A (en) 1,4-thiazine derivative
JPS62192393A (en) Dithiolindene derivative

Legal Events

Date Code Title Description
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: 2006326850

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2634212

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2008546033

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006326850

Country of ref document: AU

Date of ref document: 20061221

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2006326850

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006840387

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087017792

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200680053213.4

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2006840387

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12158563

Country of ref document: US