Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PYRIDYLUREA DERIVATIVES AND THEIR THERAPEUTIC USE
Document Type and Number:
WIPO Patent Application WO/2010/131030
Kind Code:
A1
Abstract:
Compounds of formula (IA) are inhibitors of p38 MAPK inhibitors, useful as anti-inflammatory agents in the treatment of, inter alia, diseases of the respiratory tract formula (IA). wherein R 1 is a radical of formula (IIA), (IIB), or (IIC); wherein m is 0 or 1; T is N or CH; R 2a, R 2b, R 2c are independently selected from H, halogen and C1 -C6 alkyl; R3 is H or F; R4 is -CH3; -C2 H5; -CH2OH, CH2SCH3; - SCH3 or -SC2H5; R5 is -CH3 or -C2H5; R6 is H, or represents one or more 10 substituents, each independently selected from C1 -C6 alkyl, hydroxy, halogen, and radicals of formulae (IIIA), (IIIB) and (IIIC):IA)(IIIB)(IIIC); wherein R61a and R61b are independently H or C1-C6 alkyl, or R61a and R61b taken together with the nitrogen to which they are attached to form a 6-membered 15 heterocyclic ring optionally containing a further heteroatom selected from N and O; n is 0, 1 or 2; p is 1 or 2; R7 is H or F; and R8 is H or CONH2.

Inventors:
BESWICK AMANDA (GB)
WASZKOWYCZ BOHDAN (GB)
VAN NIEL MONIQUE BODIL (GB)
Application Number:
PCT/GB2010/050767
Publication Date:
November 18, 2010
Filing Date:
May 11, 2010
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PULMAGEN THERAPEUTICS INFLAMMA (GB)
BESWICK AMANDA (GB)
WASZKOWYCZ BOHDAN (GB)
VAN NIEL MONIQUE BODIL (GB)
International Classes:
C07D213/75; C07D401/10
Domestic Patent References:
WO2002014281A12002-02-21
WO2007103468A22007-09-13
WO1999058502A11999-11-18
WO2001070695A12001-09-27
WO1998027098A11998-06-25
WO2004072038A12004-08-26
Foreign References:
US20020010170A12002-01-24
Other References:
STEIN ET AL., ANN. REP. MED CHEM., vol. 31, 1996, pages 289 - 298
HERLAAR E.; BROWN Z., MOLECULAR MEDICINE TODAY, vol. 5, 1999, pages 439 - 447
DINARELLO ET AL., REV. INFECT. DISEASE, vol. 6, 1984, pages 51
BADGER ET AL., J. PHARM. EXP. THERA., vol. 279, 1996, pages 1453 - 1461
GRISWOLD ET AL., PHARMACOL. COMM., vol. 7, 1996, pages 323 - 229
HADDAD ET AL., BR. J. PHARMACOL., vol. 132, no. 8, 2001, pages 1715 - 1724
UNDERWOOD ET AL., AM. J. PHYSIOL. LUNG CELL. MOL., vol. 279, 2000, pages 895 - 902
DUAN ET AL., AM. J. RESPIR. CRIT. CARE MED., vol. 171, 2005, pages 571 - 578
ESCOTT ET AL., BR. J. PHARMACOL., vol. 131, 2000, pages 173 - 176
UNDERWOOD ET AL., J. PHARMACOL. EXP. THER., vol. 293, 2000, pages 281 - 288
LEE ET AL., IMMUNOPHARMACOLOGY, vol. 47, 2000, pages 185 - 200
P. CHOPRA ET AL., EXPERT OPINION ON INVESTIGATIONAL DRUGS, vol. 17, no. 10, 2008, pages 1411 - 1425
G. J. HANSON, EXPERT OPINIONS ON THERAPEUTIC PATENTS, vol. 7, 1997, pages 729 - 733
J HYNES ET AL., CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 5, 2005, pages 967 - 985
C. DOMINGUEZ ET AL., EXPERT OPINIONS ON THERAPEUTICS PATENTS, vol. 15, 2005, pages 801 - 816
L. H. PETTUS; R. P. WURTZ, CURRENT TOPICS IN MEDICINAL CHEMISTRY, vol. 8, 2008, pages 1452 - 1467
EYLES JE; WILLIAMSON ED; ALPAR HO, INT J PHARM, vol. 189, no. 1, 1999, pages 75 - 9
Attorney, Agent or Firm:
PERRY, Robert, Edward (Broadgate House7 Eldon Street,London, Greater London EC2M 7LH, GB)
Download PDF:
Claims:
CLAIMS

1. A compound of formula (IA), or a pharmaceutically acceptable salt thereof;

(IA) wherein;

R1 is a radical of formula (MA), (NB), or (NC);

wherein m is 0 or 1 ;

T is N or CH;

R2a, R2b, R2c are independently selected from H, halogen and CrC6 alkyl; R3 is H or F;

R4 is -CH3, -C2H5, -CH2OH, CH2SCH3, -SCH3 or -SC2H5;

R5 is -CH3 or -C2H5;

R6 is H, or represents one or more substituents, each independently selected from CrC6 alkyl, hydroxy, halogen, and radicals of formulae (MIA), (1MB) and (NIC): * *

(HIA) (MIB) (NIC) . wherein

R61a and R61b are ind eoendentlv H or CI-CR alkvl. or R61 a and R61 b taken together with the nitrogen to which they are attached to form a 6- membered heterocyclic ring optionally containing a further heteroatom selected from N and O; n is 0, 1 or 2; p is 1 or 2; R7 is H or F; and R8 is H or CONH2.

2. A compound as claimed in claim 1 , wherein R2a, R2b and R2c are independently selected from H, F, and Cl.

3. A compound as claimed in claim 1 , wherein R2a is H, R2b is 2-chloro or 2-fluoro, and R2c is 6-chloro or 6-fluoro. 4. A compound as claimed in any of claims 1 to 3, wherein R1 is a radical of formula (MC) wherein R4 and R5 are each methyl and R6 is as defined above.

5. A compound as claimed in any of claims 1 to 3, wherein R1 is a radical of formula (MC) wherein R4 and R5 are each methyl and R6 is CrC6 alkyl, or R6 is a radical of Formula (MIB).

6. A compound as claimed in any of claims 1 to 3, wherein R1 is a radical of formula (HC) wherein R4, R5 and R6 are each methyl.

7. A compound as claimed in any of claims 1 to 3, wherein R1 is a radical of formula (MB) wherein T is CH and R3 is H. 8. A pharmaceutical composition comprising a compound as claimed in any of the preceding claims, together with one or more pharmaceutically acceptable carriers.

9. A composition as claimed in claim 8, which is adapted for inhalation for pulmonary administration.

10. A compound as claimed in any of claims 1 to 7, for use in the treatment of a disease or condition which benefits from inhibition of p38 MAP kinase activity.

11. A compound as claimed in claim 10, wherein the disease or condition is chronic eosinophilic pneumonia, asthma, COPD, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy or airways disease that is associated with pulmonary hypertension.

12. The use of a compound as claimed in any of claims 1 to 7, for the treatment of a disease or condition which benefits from inhibition of p38 MAP kinase activity.

13. The use as claimed in claim 12, wherein the disease or condition is chronic eosinophilic pneumonia, asthma, COPD, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy or airways disease that is associated with pulmonary hypertension.

Description:
PYRIDYLUREA DERIVATIVES AND THEIR THERAPEUTIC USE Field of the Invention

This invention relates to pyridylurea derivatives that are p38 MAPK inhibitors, useful as anti-inflammatory agents in the treatment of, inter alia, diseases of the respiratory tract. Background to the Invention

Mitogen-activated protein kinases (MAPK) constitute a family of proline-directed serine/threonine kinases that activate their substrates by dual phosphorylation. There are four known human isoforms of p38 MAP kinase, p38α, p38β, p38γ and p38δ. The p38 kinases, which are also known as cytokine suppressive anti-inflammatory drug binding proteins (CSBP), stress activated protein kinases (SAPK) and RK, are responsible for phosphorylating (Stein et al., Ann. Rep. Med Chem., 1996, 31 , 289-298) and activating transcription factors (such as ATF-2, MAX, CHOP and C/ERPb) as well as other kinases (such as MAPKAP-K2/3 or MK2/3), and are themselves activated by physical and chemical stress (e.g. UV, osmotic stress), proinflammatory cytokines and bacterial lipopolysaccharide (LPS) (Herlaar E. & Brown Z., Molecular Medicine Today, 1999, 5, 439-447). The products of p38 phosphorylation have been shown to mediate the production of inflammatory cytokines, including tumor necrosis factor alpha (TNFα), interleukin-1 (IL-1), and cyclooxygenase-2 (COX-2). IL-1 and TNFα are also known to stimulate the production of other proinflammatory cytokines such as IL-6 and IL-8.

IL-1 and TNFα are biological substances produced by a variety of cells, such as monocytes or macrophages. IL-1 has been demonstrated to mediate a variety of biological activities thought to be important in immunoregulation and other physiological conditions such as inflammation (e.g. Dinarello et al., Rev. Infect. Disease, 1984, 6, 51 ). Excessive or unregulated TNF production (particularly TNFα) has been implicated in mediating or exacerbating a number of diseases, and it is believed that TNF can cause or contribute to the effects of inflammation in general. IL-8 is a chemotactic factor produced by several cell types including mononuclear cells, fibroblasts, endothelial cells, and keratinocytes. Its production from endothelial cells is induced by IL-1 , TNF, or lipopolysaccharide (LPS). IL-8 stimulates a number of functions in vitro. It has been shown to have chemoattractant properties for neutrophils, T-lymphocytes and basophils. Increase in IL-8 production is also responsible for chemotaxis of neutrophils into the inflammatory site in vivo.

Inhibition of signal transduction via p38, which in addition to IL-1 , TNF and IL-8 described above is also required for the synthesis and/or action of several additional pro-inflammatory proteins (e.g., IL-6, GM-CSF, COX-2, collagenase and stromelysin), is expected to be a highly effective mechanism for regulating the excessive and destructive activation of the immune system. This expectation is supported by the potent and diverse anti-inflammatory activities described for p38 kinase inhibitors (Badger et al. , J. Pharm. Exp. Thera., 1996, 279, 1453 -1461 ; Griswold et al, Pharmacol. Comm.,1996, 7, 323-229). In particular, p38 kinase inhibitors have been described as potential agents for treating rheumatoid arthritis. In addition to the links between p38 activation and chronic inflammation and arthritis, there is also data implicating a role for p38 in the pathogenesis of airway diseases in particular COPD and asthma. Stress stimuli (including tobacco smoke, infections or oxidative products) can cause inflammation within the lung environment. Inhibitors of p38 have been shown to inhibit LPS and ovalbumin induced airway TNF-α IL-1 β, IL-6, IL-4, IL-5 and IL-13 (Haddad et al, Br. J. Pharmacol., 2001 , 132 (8), 1715-1724; Underwood et al, Am. J. Physiol. Lung Cell. MoI. 2000, 279, 895-902; Duan et al., 2005 Am. J. Respir. Crit. Care Med., 171 , 571-578; Escott et al Br. J. Pharmacol., 2000, 131 , 173- 176; Underwood et al., J. Pharmacol. Exp. Ther. 2000, 293, 281-288). Furthermore, they significantly inhibit neutrophilia and the release of MMP-9 in LPS, ozone or cigarette smoke animal models. There is also a significant body of preclinical data highlighting the potential benefits of inhibition of the p38 kinase that could be relevant in the lung (Lee et al., Immunopharmacology, 2000, 47, 185-200). Thus, therapeutic inhibition of p38 activation may be important in the regulation of airway inflammation. The implication of the p38MAPK pathway in various diseases has been reviewed by P. Chopra et al. (Expert Opinion on Investigational Drugs, 2008, 17(10), 141 1-1425). It is believed that the compounds of the present invention can be used to treat p38 mediated diseases such as: asthma, chronic or acute bronchoconstriction, bronchitis, acute lung injury and bronchiectasis, pulmonary artery hypertension, tuberculosis, lung cancer, inflammation generally (e.g. inflammatory bowel disease), arthritis, neuroinflammation, pain, fever, fibrotic diseases, pulmonary disorders and diseases (e.g., hyperoxic alveolar injury), cardiovascular diseases, post - ischemic reperfusion injury and congestive heart failure, cardiomyopathy, stroke, ischemia, reperfusion injury, renal reperfusion injury, brain edema, neurotrauma and brain trauma, neurodegenerative disorders, central nervous system disorders, liver disease and nephritis, gastrointestinal conditions, ulcerative diseases, Crohn's disease, ophthalmic diseases, ophthalmological conditions, glaucoma, acute injury to the eye tissue and ocular traumas, diabetes, diabetic nephropathy, skin-related conditions, myalgias due to infection, influenza, endotoxic shock, toxic shock syndrome, autoimmune disease, graft rejection, bone resorption diseases, multiple sclerosis, psoriasis, eczema, disorders of the female reproductive system, pathological (but non-malignant) conditions, such as hemaginomas, angiofibroma of the nasopharynx, and avascular necrosis of bone, benign and malignant tumors/neoplasia including cancer, leukaemia, lymphoma, systemic lupus erthrematosis (SLE), angiogenesis including neoplasia, haemorrhage, coagulation, radiation damage, and/or metastasis. Chronic release of active TNF can cause cachexia and anorexia, and TNF can be lethal. TNF has also been implicated in infectious diseases. These include, for example, malaria, mycobacterial infection and meningitis. These also include viral infections, such as HIV, influenza virus, and herpes virus, including herpes simplex virus type-1 (HSV-1), herpes simplex virus type-2 (HSV-2), cytomegalovirus (CMV), varicella-zoster virus (VZV), Epstein-Barr virus, human herpes virus-6 (HHV-6), human herpesvirus-7 (HHV7), human herpesvirus-8 (HHV-8), pseudorabies and rhinotracheitis, among others.

Known P38 kinase inhibitors have been reviewed by G. J. Hanson (Expert Opinions on Therapeutic Patents, 1997, 7, 729-733) J Hynes ef a/. (Current Topics in Medicinal Chemistry, 2005, 5, 967-985), C. Dominguez et al (Expert Opinions on Therapeutics Patents, 2005, 15, 801-816) and L. H. Pettus & R. P. Wurtz (Current Topics in Medicinal Chemistry, 2008, 8, 1452- 1467). P38 inhibitors containing the 1-phenyl-1-pyridin-2-yl-urea ring system are known in the art, for example W01998/027098 or WO2004/072038. Brief Description of the Invention

The compounds of the present invention are inhibitors of p38 mitogen activated protein kinase ("p38 MAPK", "p38 kinase" or "p38"), including p38α kinase, and are inhibitors of cytokine and chemokine production including TNFα and IL-8 production. They have a number of therapeutic applications, in the treatment of inflammatory diseases, particularly allergic and non- allergic airways diseases, more particularly obstructive or inflammatory airways diseases such as chronic obstructive pulmonary disease ("COPD") and asthma. They are therefore particularly suited for pulmonary delivery, by inhalation by nose or mouth. Description of the invention

According to the invention there is provided a compound of formula (IA), or a pharmaceutically acceptable salt thereof;

(IA) wherein;

R 1 is a radical of formula (HA), (MB), or (MC);

wherein m is 0 or 1 ; T is N or CH;

R 2a , R 2b , R 2c are independently selected from H, halogen and C r C 6 alkyl;

R 3 is H or F;

R 4 is -CH 3 ; -C 2 H 5 ; - CH 2 OH, CH 2 SCH 3 ; -SCH 3 or -SC 2 H 5 ; R 5 is -CH 3 or -C 2 H 5 ;

R 6 is H, or represents one or more substituents, each independently selected from C 1 -C 6 alkyl, hydroxy, halogen, and radicals of formulae (IMA), (MIB) and (NIC):

(IIIA) (IMB) (NIC) wherein

R 61a and R 61b are independently H or C r C 6 alkyl, or R 61a and R 61b taken together with the nitrogen to which they are attached to form a 6- membered heterocyclic ring optionally containing a further heteroatom selected from N and O; n is 0, 1 or 2; p is 1 or 2;

R 7 is H or F; and

R 8 is H or CONH 2 Currently preferred subclasses of compounds of the invention include: those wherein, in any compatible combination:

R 2a , R 2b and R 2c are independently selected from H, F, and Cl, for example when R 2a is H, R 2b is 2-chloro or 2-fluoro, and R 2c is 6-chloro or 6- fluoro; R 1 is a radical of formula (MC) wherein R 4 and R 5 are each methyl and

R 6 is as defined above; or

R 1 is a radical of formula (MC) wherein R 4 and R 5 are each methyl and R 6 is Ci-C 6 alkyl such as methyl, ethyl, n- or iso-propyl, or n- sec- or tert-butyl, or R 6 is a radical of Formula (MIB); or R 1 is a radical of formula (HC) wherein R 4 , R 5 and R 6 are each methyl; or

R 1 is a radical of formula (MB) wherein T is CH and R 3 is H.

In another aspect, the invention includes pharmaceutical compositions comprising a compound of the invention, together with one or more pharmaceutically acceptable carriers. Particularly preferred are compositions adapted for inhalation for pulmonary administration.

In another aspect, the invention includes the use of a compound of the invention for the treatment of diseases or conditions which benefit from inhibition of p38 MAP kinase activity. The treatment of obstructive or inflammatory airways diseases is a preferred use. All forms of obstructive or inflammatory airways diseases are potentially treatable with the compounds of the present invention, in particular an obstructive or inflammatory airways disease that is a member selected from the group consisting of chronic eosinophilic pneumonia, asthma, COPD, COPD that includes chronic bronchitis, pulmonary emphysema or dyspnea associated or not associated with COPD, COPD that is characterized by irreversible, progressive airways obstruction, adult respiratory distress syndrome (ARDS), exacerbation of airways hyper-reactivity consequent to other drug therapy and airways disease that is associated with pulmonary hypertension, chronic inflammatory diseases including cystic fibrosis, broncietasis and pulmonary fibrosis (Idiopathic). Efficacy is anticipated when p38 kinase inhibitors are administered either locally to the lung (for example by inhalation and intranasal delivery) or via systemic routes (for example, oral, intravenous and subcutaneous delivery). Terminology

As used herein, the term "(C a -C b )alkyl" wherein a and b are integers, refers to a straight or branched chain alkyl radical having from a to b carbon atoms. Thus when a is 1 and b is 6, for example, the term includes methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl and n- hexyl.

As used herein, the unqualified term "heterocyclyl" or "heterocyclic" relates to a mono-, bi- or tri-cyclic non-aromatic radical containing one or more heteroatoms selected from S, N and O, and to groups consisting of a monocyclic non-aromatic radical containing one or more such heteroatoms which is covalently linked to another such radical or to a monocyclic carbocyclic radical. Illustrative of such radicals are piperidinyl, morpholinyl, piperazinyl and N-methylpiperazinyl Compounds of the invention may exist in one or more geometrical, optical, enantiomeric, diastereomeric and tautomeric forms, including but not limited to cis- and frans-forms, E- and Z-forms, R-, S- and meso-forms, keto-, and enol-forms. Unless otherwise stated a reference to a particular compound includes all such isomeric forms, including racemic and other mixtures thereof. Where appropriate such isomers can be separated from their mixtures by the application or adaptation of known methods (e.g. chromatographic techniques and recrystallisation techniques). Where appropriate such isomers may be prepared by the application of adaptation of known methods (e.g. asymmetric synthesis).

As used herein the term "salt" includes base addition, acid addition and ammonium salts. As briefly mentioned above compounds of the invention which are acidic can form salts, including pharmaceutically acceptable salts, with bases such as alkali metal hydroxides, e.g. sodium and potassium hydroxides; alkaline earth metal hydroxides e.g. calcium, barium and magnesium hydroxides; with organic bases e.g. N-methyl-D-glucamine, choline tris(hydroxymethyl)amino-methane, L-arginine, L-lysine, N-ethyl piperidine, dibenzylamine and the like. Those compounds of the invention which are basic can form salts, including pharmaceutically acceptable salts with inorganic acids, e.g. with hydrohalic acids such as hydrochloric or hydrobromic acids, sulphuric acid, nitric acid or phosphoric acid and the like, and with organic acids e.g. with acetic, trifluoroacetic, tartaric, succinic, fumaric, maleic, malic, salicylic, citric, methanesulphonic, p-toluenesulphonic, benzoic, benzenesulfonic, glutamic, lactic, and mandelic acids and the like. Those compounds (IA) which have a basic nitrogen can also form quaternary ammonium salts with a pharmaceutically acceptable counter-ion such as, chloride, bromide, acetate, formate, p-toluenesulfonate, succinate, hemi- succinate, naphthalene-bis sulfonate, methanesulfonate, trifluoroacetate, xinafoate, and the like. For a review on salts, see Handbook of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).

It is expected that compounds of the invention may be prepared in the form of hydrates, and solvates. Any reference herein, including the claims herein, to "compounds with which the invention is concerned" or "compounds of the invention" or "the present compounds", and the like, includes reference to salts hydrates, and solvates of such compounds. The term 'solvate' is used herein to describe a molecular complex comprising the compound of the invention and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term 'hydrate' is employed when said solvent is water.

Individual compounds of the invention may exist in several polymorphic forms and may be obtained in different crystal habits.

The compounds may also be administered in the form of prodrugs thereof. Thus certain derivatives of the compounds which may be active in their own right or may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of the invention having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as 'prodrugs'. Further information on the use of prodrugs may be found in Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T. Higuchi and VJ. Stella) and Bioreversible Carriers in Drug Design, Pergamon Press, 1987 (ed. E. B. Roche, American Pharmaceutical Association; CS. Larsen and J. østergaard, Design and application of prodrugs, In Textbook of Drug Design and Discovery, 3 rd Edition, 2002, Taylor and Francis).

Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of formula (IA) with certain moieties known to those skilled in the art as 'pro- moieties' as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevier, 1985). Such examples could be a prodrug of a carboxyl group (such as -CO-O-CH 2 -O-CO-tBu as used in the pivampicillin prodrug of ampicillin), an amide (-CO-NH-CH 2 -NAIk 2 ) or an amidine (-C(=N-O-CH 3 )- NH 2 ). Utility As mentioned above the compounds of the invention are p38MAPK inhibitors, and thus may have utility for the treatment of diseases or conditions which benefit from inhibition of the p38 enzyme. Such diseases and conditions are known from the literature and several have been mentioned above. However, the compounds are generally of use as anti-inflammatory agents, particularly for use in the treatment of respiratory disease. In particular, the compounds may be used in the treatment of chronic obstructive pulmonary disease (COPD), chronic bronchitis, lung fibrosis, pneumonia, acute respiratory distress syndrome (ARDS), pulmonary emphysema, or smoking-induced emphysema, intrinsic (non-allergic asthma and extrinsic (allergic) asthma, mild asthma, moderate asthma, severe asthma, steroid resistant asthma, neutrophilic asthma, bronchitic asthma, exercise induced asthma, occupational asthma and asthma induced following bacterial infection, cystic fibrosis, pulmonary fibrosis and bronchiectasis. Compositions

As mentioned above, the compounds with which the invention is concerned are p38 kinase inhibitors, and are useful in the treatment of several diseases for example inflammatory diseases of the respiratory tract. Examples of such diseases are referred to above, and include asthma, rhinitis, allergic airway syndrome, bronchitis and chronic obstructive pulmonary disease.

It will be understood that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing treatment. Optimum dose levels and frequency of dosing will be determined by clinical trial, as is required in the pharmaceutical art. In general, the daily dose range for oral administration will lie within the range of from about 0.001 mg to about 100 mg per kg body weight of a human, often 0.01 mg to about 50 mg per kg, for example 0.1 to 10 mg per kg, in single or divided doses. In general, the daily dose range for inhaled administration will lie within the range of from about 0.1 μg to about 1 mg per kg body weight of a human, preferably 0.1 μg to 50 μg per kg, in single or divided doses. On the other hand, it may be necessary to use dosages outside these limits in some cases. For the purpose of the invention, inhaled administration is preferred.

The compounds with which the invention is concerned may be prepared for administration by any route consistent with their pharmacokinetic properties. Orally administrable compositions may be in the form of tablets, capsules, powders, granules, lozenges, liquid or gel preparations, such as oral, topical, or sterile parenteral solutions or suspensions. Tablets and capsules for oral administration may be in unit dose presentation form, and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinyl-pyrrolidone; fillers for example lactose, sugar, maize-starch, calcium phosphate, sorbitol or glycine; tabletting lubricant, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrants for example potato starch, or acceptable wetting agents such as sodium lauryl sulfate. The tablets may be coated according to methods well known in normal pharmaceutical practice. Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, glucose syrup, gelatin hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate, or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters such as glycerine, propylene glycol, or ethyl alcohol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and if desired conventional flavouring or colouring agents.

For topical application to the skin, the drug may be made up into a cream, lotion or ointment. Cream or ointment formulations which may be used for the drug are conventional formulations well known in the art, for example as described in standard textbooks of pharmaceutics such as the British Pharmacopoeia.

The active ingredient may also be administered parenterally in a sterile medium. Depending on the vehicle and concentration used, the drug can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as a local anaesthetic, preservative and buffering agents can be dissolved in the vehicle.

However, for treatment of an inflammatory disease of the respiratory tract, compounds of the invention may also be formulated for inhalation, for example as a nasal spray, or dry powder or aerosol inhalers. For delivery by inhalation, the active compound is preferably in the form of microparticles. They may be prepared by a variety of techniques, including spray-drying, freeze-drying and micronisation. Aerosol generation can be carried out using, for example, pressure-driven jet atomizers or ultrasonic atomizers, preferably using propellant-driven metered aerosols or propellant-free administration of micronized active compounds from, for example, inhalation capsules or other "dry powder" delivery systems.

By way of example, a composition of the invention may be prepared as a suspension for delivery from a nebuliser or as an aerosol in a liquid propellant, for example for use in a pressurised metered dose inhaler (PMDI). Propellants suitable for use in a PMDI are known to the skilled person, and include CFC-12, HFA-134a, HFA-227, HCFC-22 (CCI 2 F 2 ) and HFA-152 (CH 4 F 2 and isobutane)

In a preferred embodiment of the invention, a composition of the invention is in dry powder form, for delivery using a dry powder inhaler (DPI). Many types of DPI are known.

Microparticles for delivery by administration may be formulated with excipients that aid delivery and release. For example, in a dry powder formulation, microparticles may be formulated with large carrier particles that aid flow from the DPI into the lung. Suitable carrier particles are known, and include lactose particles; they may have a mass median aerodynamic diameter of greater than 90 μm.

In the case of an aerosol-based formulation, an example is: Compound of the invention 24 mg/canister Lecithin, NF Liq. Cone. 1 .2 mg/canister

Trichlorofluoromethane, NF 4.025 g/canister

Dichlorodifluoromethane, NF 12.15 g/canister

The active compounds may be dosed as described depending on the inhaler system used. In addition to the active compounds, the administration forms may additionally contain excipients, such as, for example, propellants (e.g. Frigen in the case of metered aerosols), surface-active substances, emulsifiers, stabilizers, preservatives, flavorings, fillers (e.g. lactose in the case of powder inhalers) or, if appropriate, further active compounds.

For the purposes of inhalation, a large number of systems are available with which aerosols of optimum particle size can be generated and administered, using an inhalation technique which is appropriate for the patient. In addition to the use of adaptors (spacers, expanders) and pear- shaped containers (e.g. Nebulator®, Volumatic®), and automatic devices emitting a puffer spray (Autohaler®), for metered aerosols, in particular in the case of powder inhalers, a number of technical solutions are available (e.g. Diskhaler®, Rotadisk®, Turbohaler® or the inhalers for example as described EP-A-0505321). Additionally, compounds of the invention may be delivered in multi-chamber devices thus allowing for delivery of combination agents. Combinations

Other compounds may be combined with compounds with which the invention is concerned for the prevention and treatment of inflammatory diseases, in particular respiratory diseases. Thus the present invention is also concerned with pharmaceutical compositions comprising a therapeutically effective amount of a compound of the invention and one or more other therapeutic agents. Suitable therapeutic agents for a combination therapy with compounds of the invention include, but are not limited to: (1) corticosteroids, such as fluticasone propionate, fluticasone furoate, mometasone furoate, beclometasone dipropionate, ciclesonide, budesonide, GSK 685698, GSK 870086, QAE 397, QMF 149, TPI-1020; (2) β2- adrenoreceptor agonists such as salbutamol, albuterol, terbutaline, fenoterol, and long acting β2-adrenoreceptor agonists such as salmeterol, indacaterol, formoterol (including formoterol fumarate), arformoterol, carmoterol, GSK 642444, GSK 159797, GSK 159802, GSK 597501 , GSK 678007, AZD3199; (3) corticosteroid/long acting β2 agonist combination products such as salmeterol/fluticasone propionate (Advair/Seretide), formoterol/budesonide (Symbicort), formoterol/fluticasone propionate (Flutiform), formoterol/ciclesonide, formoterol/mometasone furoate, indacaterol/mometasone furoate, Indacaterol/QAE 397, GSK 159797/GSK 685698, GSK 159802/GSK 685698, GSK 642444/GSK 685698, GSK 159797/GSK 870086, GSK 159802/GSK 870086, GSK 642444/GSK 870086, arformoterol/ciclesonide;(4) anticholinergic agents, for example muscarinic-3 (M3) receptor antagonists such as ipratropium bromide, tiotropium bromide, Aclidinium (LAS-34273), NVA-237, GSK 233705, Darotropium, GSK 573719, GSK 961081 , QAT 370, QAX 028; (5) dual pharmacology M3- anticholinergic/β2-adrenoreceptor agonists such as GSK961081 ; (6) leukotriene modulators, for example leukotriene antagonists such as montelukast, zafirulast or pranlukast or leukotriene biosynthesis inhibitors such as Zileuton or BAY-1005, or LTB4 antagonists such as Amelubant, or FLAP inhibitors such as GSK 2190914, AM-103; (7) phosphodiesterase-IV (PDE-IV) inhibitors (oral or inhaled), such as roflumilast, cilomilast, Oglemilast, ONO-6126, Tetomilast, Tofimilast, UK 500,001 , GSK 256066; (8) antihistamines, for example selective histamine-1 (H 1) receptor antagonists, such as fexofenadine, citirizine, loratidine or astemizole or dual H1/H3 receptor antagonists such as GSK 835726, GSK 1004723; (9) antitussive agents, such as codeine or dextramorphan; (10) a mucolytic, for example N acetyl cysteine or fudostein; (11) a expectorant/mucokinetic modulator, for example ambroxol, hypertonic solutions (e.g. saline or mannitol) or surfactant; (12) a peptide mucolytic, for example recombinant human deoxyribonoclease I (dornase-alfa and rhDNase) or helicidin; (13) antibiotics, for example azithromycin, tobramycin and aztreonam; (14) non-selective COX-1 / COX-2 inhibitors, such as ibuprofen or ketoprofen; (15) COX-2 inhibitors, such as celecoxib and rofecoxib; (16) VLA-4 antagonists, such as those described in WO97/03094 and WO97/02289; (17) TACE inhibitors and TNF-α inhibitors, for example anti-TNF monoclonal antibodies, such as Remicade and CDP- 870 and TNF receptor immunoglobulin molecules, such as Enbrel; (18) inhibitors of matrix metalloprotease, for example MMP-12; (19) human neutrophil elastase inhibitors, such as ONO-6818 or those described in WO2005/026124, WO2003/053930 and WO06/082412; (20) A2b antagonists such as those described in WO2002/42298; (21) modulators of chemokine receptor function, for example antagonists of CCR3 and CCR8; (22) compounds which modulate the action of other prostanoid receptors, for example a thromboxane A 2 antagonist; DP1 antagonists such as MK-0524, CRTH2 antagonists such as ODC9101 and AZD1981 and mixed DP1/CRTH2 antagonists such as AMG 009; (23) PPAR agonists including PPAR alpha agonists (such as fenofibrate), PPAR delta agonists, PPAR gamma agonists such as Pioglitazone, Rosiglitazone and Balaglitazone; (24) methylxanthines such as theophylline or aminophylline and methylxanthine/corticosteroid combinations such as theophylline/budesonide, theophylline/fluticasone propionate, theophylline/ciclesonide, theophylline/mometasone furoate and theophylline/beclometasone dipropionate; (25) A2a agonists such as those described in EP1052264 and EP1241176; (26) CXCR2 or IL-8 antagonists such as SCH 527123 or GSK 656933; (27) IL-R signalling modulators such as kineret and ACZ 885; (28) MCP-1 antagonists such as ABN-912. Methods of synthesis

Compounds of formula (I) can be prepared according to the following general scheme. Scheme 1

Compounds of general formula (l-a) may be prepared from compounds of general formula (VII):

wherein R 1 , R 2a , R 2c , R 7 and R 8 is as defined in general formula (IA), by reaction with phosgene and ammonium hydroxide.

Compounds of general formula (VII) may be prepared from compounds of general formula (V): by reaction with an amine of general formula (Vl):

R 1 NH 2 (VI), wherein R 1 is as defined in general formula (IA), using a suitable coupling agent such as 2-(7-aza-1 /-/-benzotriazole-1-yl)-1 ,1 ,3,3-tetramethyluronium hexafluorophosphate, 1 -ethyl-3-(3'-dimethylaminopropyl)carbodiimide or dicyclohexylcarbodiimide in the presence of a base such as diisopropylethylamine or triethylamine. The reaction may take place in a suitable solvent such as dichloromethane, Λ/,Λ/-dimethylformamide or tetrahydrofuran at a range of temperatures, preferably at room temperature.

Alternatively, compounds of formula (VII) may be prepared from compounds of general formula (V) by reaction with a suitable halogenating agent such as oxalyl chloride or thionyl chloride in the absence or presence of a solvent such as dichloromethane at a range of temperatures, followed by reaction with an amine of general formula (Vl), using a suitable base such as diisopropylethylamine, in a suitable solvent such as tetrahydrofuran at a range of temperatures, preferably from room temperature to 80 0 C, or by any other amide forming reaction which are well-known to those skilled in the art.

Compounds of general formula (V) may be prepared from compounds of general formula (III):

using compounds of general formula IV, where R 1 is OH.

Suitable conditions include microwave irradiation for 1-60 min, preferably 30 min and most preferably 15 min at 140 0 C using tetrakis(triphenylphosphine)palladium (0). Suitable solvents are THF and water and more preferably dioxane and water with sodium carbonate or more preferably cesium carbonate.

Compounds of general formula (IV), where R 1 is as defined for general formula IA may be prepared from compounds of general formula IV, where R 1 is OH using a suitable coupling agent such as 2-(7-aza-1 /-/-benzotriazole-1- yl)-1 ,1 ,3,3-tetramethyluronium hexafluorophosphate, 1-ethyl-3-(3'- dimethylaminopropyl)carbodiimide or dicyclohexylcarbodiimide in the presence of a base such as diisopropylethylamine or triethylamine and amine of general formula (Vl). The reaction may take place in a suitable solvent such as dichloromethane, Λ/ ; /V-dimethylformamide or tetrahydrofuran at a range of temperatures, preferably at room temperature.

Alternatively, compounds of formula (IV) where R 1 is as defined for general formula (IA) may be prepared from compounds of general formula IV, where R 1 is OH may be prepared by reaction with a suitable halogenating agent which as oxalyl chloride or thionyl chloride in the absence or presence of a solvent such as dichloromethane or Λ/,Λ/-dimethylformamide at a range of temperatures, preferably from room temperature to 100 0 C, followed by reaction with an amine of general formula (Vl), using a suitable base such as diisopropylethylamine, in a suitable solvent such as tetrahydrofuran at a range of temperatures, preferably from room temperature to 80 0 C.

Compounds of general formula (VII), where R 1 is as defined in (IA) may also be prepared from compounds of general formula (III):

by reaction with compounds of general formula (IV), where R 1 is as defined in

IA.

Suitable conditions include microwave irradiation for 1-60 min, preferably 30 min and most preferably 15 min at 140 0 C using tetrakis(triphenylphosphine)palladium (0). Suitable solvents are THF and water and more preferably dioxane and water with sodium carbonate or more preferably cesium carbonate. Compounds of general formula (III) may be prepared from compounds of general formula (I):

using compounds of general formula Il and tris(dibenzylideneacetone)dipalladium(0), and BINAP. Suitable bases include sodium terf-butoxide preferably in toluene at a range of temperatures, preferably from room temperature to 80 0 C.

General Experimental Details Abbreviations used in the experimental section: BINAP = 2,2'-

Bis(diphenylphosphino)-1 ,1 '-binaphthyl; cone = concentrated; DCM = dichloromethane; DIPEA = diisopropylethylamine; DMF = N 1 N- dimethylformamide; EtOAc = ethyl acetate; h = hours; HATU = (2-(7-aza-1H- benzotriazole-1-yl)-1 ,1 ,3,3-tetramethyluronium hexafluorophosphate); HPLC = high performance liquid chromatography; LCMS = liquid chromatography mass spectrometry; MeOH = methanol; min = minutes; RT = room temperature; Rt = retention time; THF = tetrahydrofuran

The nomenclature of structures was assigned using Autonom 2000 Name software from MDL Inc. NMR spectra were obtained on a Varian Unity Inova 400 spectrometer with a 5 mm inverse detection triple resonance probe operating at 400 MHz or on a Bruker Avance DRX 400 spectrometer with a 5 mm inverse detection triple resonance TXI probe operating at 400 MHz or on a Bruker Avance DPX 300 spectrometer with a standard 5 mm dual frequency probe operating at 300 MHz. Shifts are given in ppm relative to tetramethylsilane.

Microwave experiments were carried out using a Biotage Initiator™ Sixty, which uses a single-mode resonator and dynamic field tuning, both of which give reproducibility and control.

Purification by chromatography refers to purification using the Biotage SP1 purification system or use of the CombiFlash ® Companion purification system). Where thin layer chromatography (TLC) has been used, it refers to silica gel TLC using plates, typically 3 x 6 cm silica gel on aluminium foil plates with a fluorescent indicator (254 nm), (e.g. Fluka 60778). All solvents and commercial reagents were used as received. Compounds purified by preparative HPLC were purified using a C18-reverse-phase column (100 *

22.5 mm i.d Genesis column with 7 μm particle size, or a Phenyl-Hexyl column (250 x 21.2 mm i.d. Gemini column with 5 μm particle size, UV detection at 220, 230 or 254 nm, flow 5-15 mL/min) or Phenomenex Luna

Phenyl C6-reverse-phase column (250 * 21.20 mm 5 μm particle size) eluting with gradients from 100-0 to 0-100% water/acetonitrile or water/MeOH containing 0.1% TFA or water/acetonitrile containing 0.1% formic acid.

Detection - In-line UV detector set at 220 nM wavelength. Fractions containing the required product (identified by TLC and/or LCMS analysis) were pooled, the organic fraction removed by evaporation, and the remaining aqueous fraction lyophilised, to give the final product.

The Liquid Chromatography Mass Spectroscopy (LCMS) systems used:

Method 1

Waters Platform LC Quadrupole mass spectrometer with a C18- reverse-phase column (30 x 4.6 mm Phenomenex Luna 3 μm particle size), elution with A: water + 0.1% formic acid; B: methanol + 0.1% formic acid.

Gradient:

Gradient - Time flow mL/min %A %B

0.00 2.0 95 5 0.50 2.0 95 5

4.50 2.0 5 95

5.50 2.0 5 95

6.00 2.0 95 5

Detection - MS, ELS, UV (200 μL split to MS with in-line UV detector). MS ionization method - Electrospray (positive and negative ion).

Method 2

Waters Micromass ZQ2000 with a C18-reverse-phase column (100 *

3.0 mm Higgins Clipeus with 5 μm particle size), elution with A: water + 0.1% formic acid; B: acetonitrile + 0.1% formic acid. Gradient: Gradient - Time flow mL/min %A %B

0.00 1.0 95 5

1.00 1.0 95 5

15.00 1.0 5 95 20.00 1.0 5 95

22.00 1.0 95 5

25.00 1.0 95 5

Detection - MS, ELS, UV (100 μL split to MS with in-line UV detector). MS ionisation method - Electrospray (positive ion) Method 3

Waters ZMD quadrupole mass spectrometer with a C18-reverse- phase column (30 * 4.6 mm Phenomenex Luna 3 μm particle size), elution with A: water + 0.1 % formic acid; B: methanol + 0.1 % formic acid. Gradient:

Gradient - Time flow mL/min %A %B 0.00 2.0 95 5

0.50 2.0 95 5

4.50 2.0 5 95

5.50 2.0 5 95

6.00 2.0 95 5 Detection - MS, ELS, UV (200 μL split to MS with in-line Waters 996 DAD detector). MS ionization method - Electrospray (positive and negative ion).

Method 4

Waters Micromass ZQ2000 with a C18-reverse-phase column (100 x

3.0 mm Higgins Clipeus with 5 μm particle size), elution with A: water + 0.1 % formic acid; B: methanol + 0.1 % formic acid. Gradient:

Gradient - Time flow mL/min %A %B

0.00 1.0 85 15

1.00 1.0 85 15

15.00 1.0 5 95 20.00 1.0 5 95

22.00 1.0 85 15

25.00 1.0 85 15

Detection - MS, ELS, UV (100 μL split to MS with in-line UV detector). MS ionisation method - Electrospray (positive/negative ion). Method 5

Waters Quatro Microtriple quadrupole with a C18-reverse-phase column (100 x 3.0 mm Higgins Clipeus with 5 μm particle size), elution with A: water + 0.1 % formic acid; B: methanol + 0.1 % formic acid. Gradient: Gradient - Time flow mL/min %A %B

0.00 1.0 85 15

1.00 1.0 85 15

13.00 1.0 5 95

20.00 1.0 5 95 22.00 1.0 85 15

25.00 1.0 85 15

Detection - MS, ELS, UV (100 μl_ split to MS with in-line UV detector). MS ionisation method - Electrospray (positive/negative ion).

Example 1 W-(5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-(3-{6-[1 -(2,6-difluoro-phenyl)- ureido]-pyridin-2-yl}-phenyl)-acetamide

a. (6-Chloro-pyridin-2-yl)-(2,6-difluoro-phenyl)-amine

A solution of 2-bromo-6-chloro-pyridine (3.0 g, 15.6 mmol), 2,6- difluoroaniline (1.76 ml_, 16.37 mmol), tris(dibenzylideneacetone)dipalladium(0) (285 mg, 0.31 mmol), BINAP (388 mg, 0.62 mmol) and sodium fe/f-butoxide (2.1 g, 21.83 mmol) in toluene (50 ml_) was purged with argon, stirred at 70 0 C for 7 h then allowed to cool down to RT. The reaction mixture was partitioned between diethyl ether and water. The aqueous layer was extracted twice with diethyl ether, the combined organic layers were washed with brine, dried (MgSO 4 ), concentrated in vacuo and purified twice by chromatography (0 to 10% ethyl acetate in cyclohexane then 0 to 30% DCM in cyclohexane) to give the title compound as a white solid (1.78 g, 47%). LCMS (Method 1 ): Rt 4.55 min, m/z 241/243 [MH + ]. 1 H NMR (400 MHz, DMSO-d 6 ): 8.90 (1 H, s), 7.57 (1 H, dd, J 8.2, 7.5), 7.34-7.25 (1 H, m), 7.21-7.13 (2 H, m), 6.77 (1 H, dd, J 7.5, 0.6), 6.63 (1 H, d, J 8.2). b. (5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-[3-(4,4,5,5-tetra methyl-

[1 ,3,2]dioxaborolan-2-yl)-phenyl]-acetamide

A solution of 5-tert-butyl-2-p-tolyl-2H-pyrazol-3-ylamine hydrochloride (J.Med. Chem., 2002, 45, 2994) (200 mg, 0.75 mmol) in DCM (3 ml.) and

DMF (1 mL) was treated with [3-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)- phenyl]-acetic acid (217 mg, 0.83 mmol), DIPEA (0.29 mL, 0.65 mmol), and

HATU (316 mg, 0.83 mmol). The reaction mixture was stirred at RT for 48 h, then partitioned between DCM and water. The aqueous layer was extracted twice with DCM, the combined organic layers were washed with water, brine, dried (MgSO 4 ), concentrated in vacuo and purified by chromatography (0 to

30% EtOAc in cyclohexane) to give the title compound as a white foam

(190 mg, 53%). LCMS (Method 1 ): Rt 4.89 min, m/z 474 [MH + ]. 1 H NMR (400

MHz, DMSO-de): 9.99 (1 H, s), 7.62 (1 H, s), 7.57 (1 H, d, J 7.1 ), 7.40-7.30 (2 H, m), 7.28-7.22 (2 H, m), 7.18 (2 H, d, J 8.2), 6.27 (1 H, s), 3.58 (2 H, s),

2.33 (3 H, s), 1.30 (12 H, s), 1.25 (9 H, s). c. Λ/-(5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-{3-[6-(2,6-di fluoro- phenylamino)-pyridin-2-yl]-phenyl}-acetamide

A solution of Example 1 step a (86 mg, 0.36 mmol), Example 1 step b

(187 mg, 0.39 mmol), tetrakis(triphenylphosphine)palladium(0) (42 mg, 0.036 mmol) and cesium carbonate (234 mg, 0.72 mmol) in dioxane (4 ml_) and water (2 ml_) was purged with argon and heated in a microwave at 140°C for 15 min. The reaction mixture was partitioned between diethyl ether and water, the aqueous layer was extracted twice with diethyl ether, the combined organic layers were washed with brine, dried (MgSO 4 ), concentrated in vacuo and purified by chromatography (0 to 20% EtOAc in cyclohexane) to give the title compound as a white solid (168 mg, 78%).

LCMS (Method 2): Rt 13.42 min, m/z 552 [MH + ]. 1 H NMR (400 MHz, DMSO- (J 6 ): 9.95 (1 H, s), 8.64 (1 H, s), 7.76 (2 H, m), 7.65 (1 H, t, J 7.8), 7.33 (1 H, t, J 7.6), 7.26-7.17 (5 H, m), 7.19-7.09 (2 H, m), 7.08 (2 H, d, J 8.1 ), 6.69 (1 H, d, J 8.2), 6.27 (1 H, s), 3.60 (2 H, s), 2.23 (3 H, s), 1.25 (9 H, s). d. Λ/-(5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-(3-{6-[1-(2,6 -difluoro- phenyl)-ureido]-pyridin-2-yl}-phenyl)-acetamide

A solution of Example 1 step c (87 mg, 0.16 mmol) in dry toluene (4 ml_) under nitrogen was treated with a 20% phosgene solution in toluene (0.33 ml_, 0.63 mmol), and heated at 55°C for 48 h. The reaction mixture was cooled to -5°C, treated with aqueous 33% ammonium hydroxide solution (4 mL), allowed to warm up to RT, sonicated, diluted with water and stirred at RT for 1 h to give a white suspension. The residue was filtered, purified by chromatography (0 to 40% EtOAc in cyclohexane), triturated with diethyl ether, filtered and dried to give the title compound as a white solid (35 mg, 37%). LCMS (Method 2): Rt 12.12 min, m/z 595 [MH + ]. 1 H NMR (400 MHz, DMSO-d 6 ): 9.96 (1 H, s), 7.85 (1 H, t, J 8.0), 7.75-7.67 (3 H, m), 7.60-7.46 (2 H, m), 7.40 (1 H, t, J 7.6), 7.32-7.17 (6 H, m), 7.07 (2 H, d, J 8.1 ), 7.04 (1 H, d, J 8.4), 6.28 (1 H, s), 3.62 (2 H, s), 2.22 (3 H, s), 1.26 (9 H, s). Example 2

Λ/-{5-fert-Butyl-2-[4-(2-morpholin-4-yl-ethoxy)-phenyl]- 2H-pyrazol-3-yl}-2- (3-{6-[1-(2,6-difluoro-phenyl)-ureido]-pyridin-2-yl}-phenyl) -acetamide

a. 5-fert-Butyl-2-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-2H-pyraz ol-3- ylamine

A solution of 4-(5-amino-3-te/t-butyl-pyrazol-1-yl)-phenol (462 mg, 2 mmol) (WO2007/059202), 2-morpholin-4-yl-ethanol (327 mg, 2.5 mmol) and triphenylphosphine (1.05 g, 4 mmol) in dry THF (5 ml_) was treated dropwise with diisopropyl azodicarboxylate (808 mg, 4 mmol). The reaction mixture was stirred at RT for 16 h, then partitioned between diethyl ether and water. The aqueous layer was basified with K 2 CO 3 and extracted twice with EtOAc. The combined organic layers were dried (Na 2 SO 4 ), concentrated in vacuo and triturated with diethyl ether to give the title compound as a white solid (297 mg, 43%). LCMS (Method 1 ): Rt 2.15 min, m/z 345 [MH + ]. 1 H NMR (400 MHz, CDCI 3 ): 7.44-7.39 (2 H, m), 6.97-6.92 (2 H, m), 5.48 (1 H, s), 4.14- 4.07 (2 H, m), 3.73 (4 H, t, J 4.6 Hz), 3.61 (2 H, s), 2.82-2.77 (2 H, m), 2.58 (4 H, m), 1.33-1.27 (9 H, s). b. Λ/-{5-te/t-Butyl-2-[4-(2-morpholin-4-yl-ethoxy)-phenyl]-2H- pyrazol-3- yl}-2-(3-{6-[1-(2,6-difluoro-phenyl)-ureido]-pyridin-2-yl}-p henyl)- acetamide

The title compound was prepared in a similar manner to that described for Example 1 steps b-d using 2 step a. LCMS (Method 4): Rt 9.22 min, m/z 710 [MH + ]. 1 H NMR (400 MHz, CD 3 OD): 7.85-7.75 (3 H, m), 7.57-7.44 (2 H, m), 7.41 (1 H, t, J 7.7 Hz), 7.29 (1 H, d, J 7.7 Hz), 7.23-7.03 (4 H, m), 6.78- 6.64 (3 H, m), 6.36 (1 H, s), 3.97 (2 H, t, J 5.4 Hz), 3.75-3.62 (4 H, m), 3.62 (2 H, s) 2.74 (2 H, t, J 5.4 Hz), 2.58 (4 H, t, J 4.4 Hz), 1.32 (9 H, s). Example 3

Λ/-(5-fe/t-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-(3-{6-[1-( 2,6-difluoro-phenyl)- ureido]-pyridin-2-yi}-4-fluoro-phenyl)-acetamide a. [4-Fluoro-3-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-phenyl]- acetic acid

A suspension of (3-bromo-4-fluoro-phenyl)-acetic acid (1g,

4.29 mmol), bis (pinacolato)diboron (1.3 g, 5.15 mmol) and KOAc (1.28 g, 13 mmol) in dioxane (15 mL) was purged with argon and treated with dichlorobis(diphenylphospino)palladium. The reaction mixture was heated at 90 0 C for 24 h, diluted with EtOAc and extracted with sat. solution of NaHCO 3 . The aqueous layer was acidified with cone. HCI and extracted twice with EtOAc. The combined organic layers were dried (Na 2 SO 4 ), concentrated in vacuo, and purified by chromatography (0 to 100% EtOAc in cyclohexane with 1 % acetic acid) to give the title compound as a white solid (520 mg, 43%). LCMS (Method 3): Rt 2.66 min, m/z 281 [MH + ]. 1 H NMR (300 MHz, CDCI 3 ): 7.61 (1 H, dd, J 5.5, 2.5), 7.35 (1 H, ddd, J 8.5, 5.2, 2.5), 7.00 (1 H, t, J 8.7), 3.62 (2 H, s), 1.35 (12 H, s). b. Λ/-(5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-yl)-2-(3-{6-[1-(2,6 -difluoro- phenyl)-ureido]-pyridin-2-yl}-4-fluoro-phenyl)-acetamide

The title compound was prepared in a similar manner to that described for Example 1 using 3 step a, and performing the steps in the order c, b, d. LCMS (Method 4): Rt 12.09 min, m/z 613 [MH + ]. 1 H NMR (400 MHz, CDCI 3 ): 7.68 (1 H, t, J 8.1 Hz), 7.58 (1 H, dd, J 7.2, 2.3), 7.42-7.33 (2 H, m), 7.25-7.20 (2 H, m), 7.14 (1 H, dd, J 10.9, 8.4 Hz), 7.09-6.95 (5 H, m), 6.72 (1 H, d, J 8.5), 6.63 (1 H, s), 3.71 (2 H, s), 2.24 (3 H, s), 1.34 (9 H, s). Example 4

2-{3-[(5-fert-Butyl-2-p-tolyl-2H-pyrazol-3-ylcarbamoyl)-m ethyl]-phenyl}-6- [1-(2,6-difluoro-phenyl)-ureido]-nicotinamide a. β-Bromo^-chloro-nicotinic acid ethyl ester

A solution of 2-chloro-1-oxide-3-pyridinecarboxylic acid ethyl ester (W01995/005378) (1.21 g, 6 mmol) and phosphorus (V) oxybromide (3.45 g,

12 mmol) in acetonitrile (25 mL) was heated at 70 0 C for 18h, then quenched with water. The aqueous layer was extracted with DCM and the resulting organic layer was washed with sat. sodium bicarbonate solution, dried

(MgSO 4 ), filtered, concentrated in vacuo and purified by chromatography (cyclohexane/EtOAc 100/0 to 75/25) to afford the title compound as a colourless solid (0.37 g, 23%). 1 H NMR (300 MHz, CDCI 3 ): 7.92 (1 H, d, J 8),

7.53 (1 H, d, J 8), 4.42 (2 H, q, J 7.1 ), 4.42 (3 H, t, J 7.1 ). b. 2-Chloro-6-(2,6-difluoro-phenylamino)-nicotinic acid ethyl ester

The title compound was prepared in a similar manner to that described for Example 1 step a using 4 step a. The compound was obtained as a 1 :0.35 mixture with 6-bromo-2-(2,6-difluoro-phenylamino)-nicotinic acid ethyl ester. LCMS (Method 3): Rt 4.17 min, m/z 313 and 357/359 [MH + ]. c. 2-{3-[(5-tert-Butyl-2-p-tolyl-2H-pyrazol-3-ylcarbamoyl)-meth yl]-phenyl}- 6-(2,6-difluoro-phenylamino)-nicotinic acid ethyl ester

The title compound was prepared in a similar manner to that described for Example 1 step c using 4 step b. LCMS (Method 3): Rt 4.70 min, m/z 624 [MH + ]. d. 2-{3-[(5-fe/t-Butyl-2-p-tolyl-2H-pyrazol-3-ylcarbamoyl)-meth yl]- phenyl}-6-[1 -(2,6-difluoro-phenyl)-ureido]-nicotinamide

A solution of 4 step c (70 mg, 0.112 mmol) and 1 N sodium hydroxide (0.22 ml_, 0.22 mmol) in THF (2 ml_) was heated to 55°C for 4h. More 1 N sodium hydroxide (0.44 ml_, 0.44 mmol) was added and the reaction mixture was heated at 70°C for 16h. More 1 N sodium hydroxide (1 ml_, 1 mmol), solid sodium hydroxide (600 mg, 15 mmol) and methanol (2 ml_) were added and heating was pursued at 80 0 C for 4h. The reaction mixture was cooled, acidified with cone. HCI and extracted with DCM. The resulting organic layer was washed with brine, dried (MgSO 4 ), filtered and concentrated in vacuo. The resulting oil was dissolved in dry toluene (4 ml_), treated with phosgene (20% solution in toluene, 0.35 ml_, 6.74 mmol) and heated at 55°C for 2Oh. The reaction mixture was concentrated in vacuo, dissolved in dry THF (2 ml_), treated with 33% aq. ammonia solution (1 ml.) and toluene (1 ml_) and stirred at RT for 3h. The resulting reaction mixture was extracted with DCM, dried (MgSO 4 ), filtered, concentrated in vacuo and purified by chromatography (cyclohexane/EtOAc 100/0 to 0/100) to afford the title compound as a pale yellow solid (8 mg, 1 1 %). LCMS (Method 5): Rt 10.43 min, m/z 638 [MH + ]. 1 H NMR (400 MHz, DMSO-d 6 ): 9.99 (1 H, s), 7.81 (2 H, m), 7.54-7.44 (4 H, m), 7.39-7.21 (8 H, m), 7.13 (2 H, d, J 8.2), 6.98 (1 H, d, J 8.5), 6.30 (1 H, s), 3.58 (2 H, s), 2.27 (3 H, s), 1.26 (9 H, s). Example 5

2-(3-{6-[1-(2,6-Difluoro-phenyl)-ureido]-pyridin-2-yl}-ph enyl)-N-(3- morpholin-4ylphenyl)-acetamide

The title compound was prepared using similar methods to that described for the preparation of Example 1 (Method 4): Rt 10.62 min, m/z 544

[MH + ]. (DMSO-d 6 ):10.05 (1 H, s), 7.84 (1 H, t, J 8.0), 7.78 (1 H, s), 7.69 (1 H, dt, J 7.2, 1.8), 7.62 (1 H, d, J 7.7), 7.55-7.44 (1 H, m), 7.43-7.35 (2 H, m), 7.30-7.22 (3 H, m), 7.14 (1 H, t, J 8.1 ), 7.04 (2 H, m), 6.65 (1 H, dd, J 8.2, 2.4), 3.77 (4 H, m), 3.64 (2 H, s), 3.05 (4 H, t, J 4.7). Biological assays p38 Kinase Assay

Human recombinant p38 enzyme expressed in E. coli and activated by incubation with MKK6 enzyme (Calbiochem #559324) is used as source of enzyme activity.

The assay is carried in high binding, clear, flat bottom 96 well assay plates which have been coated with recombinant ATF-2 (Biosource #PHF0043). Test compounds are incubated with p38 kinase for 2h prior to initiating the kinase assay by the addition of ATP to obtain an assay concentration of 250 μM. Phosphorylation of ATF-2 is detected and quantified using an ELISA. This consists of sequential incubation in the presence of anti-phospho-ATF2, biotinylated anti-lgG and streptavidin-HRP. Incubation with an HRP chromogenic substrate (TMB) results in absorbance that is proportional to the amount of phosphorylated substrate produced. Absorbance is detected using a multiwell plate reader.

Compounds are diluted in DMSO prior to addition to assay buffer, the final DMSO concentration in the assay being 1 %.

The IC 50 is defined as the concentration at which a given compound achieves 50% inhibition of control.

Table 1

In the table above, p38α binding potencies (IC 50 values) are indicated as follows: <1000nM '+'; <500nM '++'; <100nM '+++'; <10nM '++++'. All compounds tested exhibited IC 50 values <1000nM. NT = Not Tested. p38 functional assay Inhibition of cellular p38 depresses the release of TNFα, a functional response which is quantified by measurement of the amount of TNFα in the supematants of LPS activated THP- 1 cells (an immortalised monocytic cell line) or peripheral blood mononuclear cells (PBMC's) isolated from freshly drawn human blood.

Cells seeded in 96 well plates are pre-treated by the addition of p38 inhibitors for 1 h followed by addition of lipopolysaccharide (LPS) to activate cytokine production and release. The amount of TNFα released into the cell supematants is quantified using an R&D Systems enzyme linked immunosorbant assay (ELISA) kit (product DY210) following the manufacturers instructions.

Compounds are diluted in DMSO prior to addition, the final DMSO concentration in the assay being 0.3%. The EC 5O is defined as the concentration at which a given compound achieves 50% inhibition of the control. Results are shown in Table 2:

Table 2

In Table 2 above, EC 5O values are indicated as follows: <7000-500nM '+'; <500-100nM '++'; 10 -<100nM '+++'; <10nM '++++'. All compounds tested exhibited EC 50 values <2000nM; NT not tested.

Pre-clinical mouse model of COPD inflammation - Tobacco smoke induced pulmonary inflammation.

Previous studies have established that the number of inflammatory cells recovered in the bronchoalveolar lavage (BAL) is significantly elevated 24 h following the final Tobacco Smoke (TS) exposure of 4 or 1 1 consecutive daily TS exposures, this time point was used in the studies reported here. Protocols for the exposure of mice to TS, obtaining bronchoalveolar lavage (BAL), preparation of cytospin slides for differential cell counts are as outlined below.

Exposure of mice to TS daily for 4 or 11 consecutive days In this exposure protocol, mice were exposed in groups of 5 in individual clear polycarbonate chambers (27 cm x 16 cm x 12 cm). The TS from the cigarettes was allowed to enter the exposure chambers at a flow rate of 100 ml/min. In order to minimise any potential problems caused by repeated exposure to a high level of TS (6 cigarettes), the exposure of the mice to TS was increased gradually over the exposure period to a maximum of 6 cigarettes. The exposure schedule used for 4 days was as follows:

Day 1 : 4 cigarettes (approximately 32 min exposure)

Day 2: 4 cigarettes (approximately 32 min exposure)

Day 3: 6 cigarettes (approximately 48 min exposure) Day 4: 6 cigarettes (approximately 48 min exposure)

The exposure schedule used for 11 days exposure was as follows: Day 1 : 2 cigarettes (approximately 16 min exposure)

Day 2: 3 cigarettes (approximately 24 min exposure)

Day 3: 4 cigarettes (approximately 32 min exposure) Day 4: 5 cigarettes (approximately 40 min exposure)

Day 5 to 1 1 : 6 cigarettes (approximately 48 min exposure) A further group of mice were exposed to air on a daily basis for equivalent lengths of time as controls (no TS exposure). Bronchoalveolar lavage (BAL) analysis Bronchoalveolar lavage was performed as follows: the trachea was cannulated using a Portex nylon intravenous cannula (pink luer fitting) shortened to approximately 8 mm. Phosphate buffered saline (PBS) was used as the lavage fluid. A volume of 0.4 ml was gently instilled and withdrawn 3 times using a 1 ml syringe and then placed in an Eppendorf tube and kept on ice prior to subsequent determinations. Cell counts:

Lavage fluid was separated from cells by centrifugation and the supernatant decanted and frozen for subsequent analysis. The cell pellet was re-suspended in a known volume of PBS and total cell numbers calculated by counting a stained (Turks stain) aliquot under a microscope using a haemocytometer.

Differential cell counts were performed as follows:

The residual cell pellet was diluted to approximately 10 5 cells per ml. A volume of 500 μl was placed in the funnel of a cytospin slide and centrifuged for 8 min at 800 rpm. The slide was air dried and stained using

'Kwik-Diff solutions (Shandon) as per the proprietary instructions. When dried and cover-slipped, differential cells were counted using light microscopy. Up to 400 cells were counted by unbiased operator using light microscopy. Cells were differentiated using standard morphometric techniques. Drug Treatment

Rodents such as mice and rats are obligate nasal breathers thus oral delivery of test materials (such as therapeutic agents) for inhalation will not produce good lung exposure. As a consequence, delivery of therapeutic agents to the lungs in rodents is generally achieved by intra-nasal, intratracheal or inhalation by whole body aerosol exposure in a chamber.

The chamber method utilises large amounts of test material and is generally reserved for inhalation toxicology studies rather than pharmacological efficacy studies. Intra-tracheal administration is a very efficient delivery method as almost all of the test material is delivered to the lungs, but this is quite an invasive technique. For studies in the mouse particularly, it is also quite technically demanding as the diameter of the trachea is quite small. The intranasal route is less invasive than the intra- tracheal route and so is particularly suitable for repeat dosing studies such as the 4-1 1 day mouse model described below. Following intranasal administration ~50% of the dose administered is delivered to the lungs (Eyles JE, Williamson ED and Alpar HO. 1999, lnt J Pharm, 189(1 ):75-9).

As a surrogate route for oral inhalation, mice were dosed intra-nasally with vehicle (0.2% tween 80 in saline) and Example 1 (100 μg/kg). The control group of mice received vehicle 1 hr prior to being exposed to air daily for a maximum of 50 minutes per day. TS exposure was conducted for 4 days. BAL was performed 24 h following the final TS exposure. Data management and statistical analysis All results are presented as individual data points for each animal and the mean value was calculated for each group. Since tests for normality were positive, the data were subjected to a one way analysis of variance test (ANOVA), followed by a Bonferroni correction for multiple comparisons in order to test for significance between treatment groups. A "p" value of < 0.05 was considered to be statistically significant. Percentage inhibitions were automatically calculated within the Excel spreadsheets for the cell data using the formula below:

% Inhibition = 1 - x 100 TS vehicle group result - sham group result J Inhibition data for other parameters were calculated manually using the above formula.

Brief description of the Drawings

Figure 1 is a bar graph that illustrates the effect of intranasal administration to laboratory mice with vehicle (0.2% tween 80 in saline) or Example 1 (100 μg/kg) on the number of BAL cells induced by tobacco smoke

24 hours post the final exposure.

Figure 2 is a bar graph that illustrates the effect of intranasal administration to laboratory mice with vehicle (0.2% tween 80 in saline) or

Example 1 (100 μg/kg), on the number of BAL neutrophils induced by tobacco smoke 24 hours post the final exposure.

As illustrated in Figure 1 , Example 1 significantly inhibited the BAL cell influx induced by TS at 100μg/kg when administered by the intranasal route.

Similar findings were observed with BAL neutrophils (Figure 2). The results demonstrate a clear anti-inflammatory effect in the lungs of mice exposed to tobacco smoke (TS).