US20150225373A1 - Kinase inhibitors - Google Patents

Kinase inhibitors Download PDF

Info

Publication number
US20150225373A1
US20150225373A1 US14/424,627 US201314424627A US2015225373A1 US 20150225373 A1 US20150225373 A1 US 20150225373A1 US 201314424627 A US201314424627 A US 201314424627A US 2015225373 A1 US2015225373 A1 US 2015225373A1
Authority
US
United States
Prior art keywords
compound
alkyl
formula
group
halo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/424,627
Other languages
English (en)
Inventor
Matthew Colin Thor Fyfe
Premji Meghani
Matthew Alexander Henry Stent
Stephen Malcolm Thom
Claire Anne Walshe
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Respivert Ltd
TopiVert Pharma Ltd
Original Assignee
Respivert Ltd
TopiVert Pharma 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
Priority claimed from GBGB1215369.8A external-priority patent/GB201215369D0/en
Priority claimed from GB201304777A external-priority patent/GB201304777D0/en
Application filed by Respivert Ltd, TopiVert Pharma Ltd filed Critical Respivert Ltd
Assigned to TOPIVERT PHARMA LIMITED, RESPIVERT LIMITED reassignment TOPIVERT PHARMA LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THOM, Stephen Malcolm, MEGHANI, PREMJI, FYFE, MATTHEW COLIN THOR, WALSHE, CLAIRE ANN, STENT, MATTHEW ALEXANDER HENRY
Publication of US20150225373A1 publication Critical patent/US20150225373A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/01Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms
    • C07C255/17Carboxylic acid nitriles having cyano groups bound to acyclic carbon atoms containing cyano groups and doubly-bound oxygen atoms bound to the same acyclic carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C51/00Preparation of carboxylic acids or their salts, halides or anhydrides
    • C07C51/347Preparation of carboxylic acids or their salts, halides or anhydrides by reactions not involving formation of carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/0825Preparations of compounds not comprising Si-Si or Si-cyano linkages
    • C07F7/083Syntheses without formation of a Si-C bond

Definitions

  • This invention relates, inter alia, to compounds which are antiinflammatory agents (e.g. through inhibition of one or more of members of: the family of p38 mitogen-activated protein kinase enzymes (referred to herein as p38 MAP kinase inhibitors), for example the alpha kinase sub-type thereof; Syk kinase; and the Src family of tyrosine kinases).
  • p38 mitogen-activated protein kinase enzymes referred to herein as p38 mitogen-activated protein kinase enzymes (referred to herein as p38 MAP kinase inhibitors), for example the alpha kinase sub-type thereof; Syk kinase; and the Src family of tyrosine kinases).
  • the invention also relates to the use of such compounds in therapy, including in mono- and combination therapies, especially in the treatment of inflammatory diseases, including inflammatory diseases of the lung (such as asthma and chronic obstructive pulmonary disease (COPD)), eye (such as uveitis) and gastrointestinal tract (such as Crohn's disease and ulcerative colitis).
  • inflammatory diseases including inflammatory diseases of the lung (such as asthma and chronic obstructive pulmonary disease (COPD)), eye (such as uveitis) and gastrointestinal tract (such as Crohn's disease and ulcerative colitis).
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • eye such as uveitis
  • gastrointestinal tract such as Crohn's disease and ulcerative colitis
  • p38 MAPK isoforms (alpha, beta, gamma and delta respectively) have been identified, each displaying different patterns of tissue expression.
  • the p38 MAPK alpha and beta isoforms are found ubiquitously throughout the body; are present in many different cell types and are inhibited by a number of previously described small molecular weight compounds. Early classes of inhibitors were highly toxic due to the broad tissue distribution of these isoforms which resulted in off-target effects of the compounds. Some of the more recently identified inhibitors show improved selectivity for p38 MAPK alpha and beta isoforms and have wider safety margins.
  • p38 MAP kinase is believed to play a pivotal role in many of the signalling pathways that are involved in initiating and maintaining chronic, persistent inflammation in human disease, for example, in severe asthma, COPD and inflammatory bowel disease (IBD).
  • IBD inflammatory bowel disease
  • p38 MAP kinase is activated by a range of pro-inflammatory cytokines and that its activation results in the recruitment and release of further pro-inflammatory cytokines.
  • data from some clinical studies demonstrate beneficial changes in disease activity in patients during treatment with p38 MAP kinase inhibitors. For instance Smith describes the inhibitory effect of p38 MAP kinase inhibitors on TNF ⁇ (but not IL-8) release from human PBMCs (Smith, S. J., Br. J. Pharmacol., 2006, 149:393-404).
  • COPD is a condition in which the underlying inflammation is reported to be substantially resistant to the anti-inflammatory effects of inhaled corticosteroids. Consequently, a superior strategy for treating COPD would be to develop an intervention which has both inherent anti-inflammatory effects and the ability to increase the sensitivity of the lung tissues of COPD patients to inhaled corticosteroids.
  • the recent publication of Mercado et al. (2007 ; American Thoracic Society Abstract A 56) demonstrates that silencing p38 MAPK gamma has the potential to restore sensitivity to corticosteroids. Thus, there may be a dual benefit for patients in the use of a p38 MAP kinase inhibitor for the treatment of COPD.
  • Certain p38 MAPK inhibitors have also been described as inhibitors of replication of respiratory syncytial virus (Cass L. et al., WO 2011/158039).
  • IBD The precise etiology of IBD is uncertain, but is believed to be governed by genetic and environmental factors that interact to promote an excessive and poorly controlled mucosal inflammatory response directed against components of the luminal microflora. This response is mediated through infiltration of inflammatory neutrophils, dendritic cells and T-cells from the periphery. Due to the ubiquitous expression of p38 in inflammatory cells it has become an obvious target for investigation in IBD models. Studies investigating the efficacy of p38 inhibitors in animal models of IBD and human biopsies from IBD patients indicated that p38 could be a target for the treatment of IBD (Hove, T. ten et al., Gut, 2002, 50:507-512, Docena, G.
  • T cells are known to play key role in mediating inflammation of the gastrointestinal tract.
  • SCID immunodeficient
  • mucosal membranes from IBD patients showed an upregulation of CD4+ cells which were either Th1 (IFN ⁇ /IL-2) or Th2 (IL5/TGF ⁇ ) biased depending on whether the patient had Crohn's disease or ulcerative colitis (Fuss I J. et al. J Immunol. 1996 157:1261-70.).
  • T cells are known to play a key role in inflammatory disorders of the eye with several studies reporting increased levels of T cell associated cytokines (IL-17 and IL-23) in sera of Behçets patients (Chi W. et al. Invest Ophthalmol Vis Sci. 2008 49:3058-64). In support, Direskeneli and colleagues demonstrated that Behçets patients have increased Th17 cells and decreased Treg cells in their peripheral blood (Direskeneli H. et al. J Allergy Clin Immunol. 2011 128:665-6).
  • Src family kinases are known to play a key role in this pathway, where Src family kinases, Fyn and Lck, are the first signalling molecules to be activated downstream of the T cell receptor (Barber E K. et al. PNAS 1989 86:3277-81). They initiate the tyrosine phosphorylation of the T cell receptor leading to the recruitment of the Syk family kinase, ZAP-70. Animal studies have shown that ZAP-70 knockout results in a SCID phenotype (Chan A C. et al. Science. 1994 10; 264(5165): 1599-601).
  • Syk kinase is widely expressed in cells of the hematopoietic system, most notably in B cells and mature T cells. Through interaction with immunoreceptor tyrosine-based activation (ITAM) motifs it plays an important role in regulating T cell and B cell expansion as well as mediating immune-receptor signalling in inflammatory cells.
  • ITAM immunoreceptor tyrosine-based activation
  • IL-6 and MMP release inflammatory mediators commonly found upregulated in inflammatory disorders including IBD and rheumatoid arthritis (Wang Y D. et at World J Gastroenterol 2007; 13: 5926-5932, Litinsky I et al. Cytokine. 2006 January 33:106-10).
  • kinase enzymes are now also recognised to regulate the activity of a range of cellular functions, including the maintenance of DNA integrity (Shilo, Y. Nature Reviews Cancer, 2003, 3: 155-168) and co-ordination of the complex processes of cell division. Indeed, certain kinase inhibitors (the so-called “Olaharsky kinases”) have been found to alter the frequency of micronucleus formation in vitro (Olaharsky, A. J. et al., PLoS Comput. Biol., 2009, 5(7)). Micronucleus formation is implicated in, or associated with, disruption of mitotic processes and is therefore undesirable.
  • GSK3 ⁇ glycogen synthase kinase 3 ⁇
  • urea derivatives are disclosed as having anti-inflammatory properties (see, for example, WO 01/36403, WO 01/4115, WO 02/092576, WO 2003/068228, WO 2003/072569, WO 2004/113352, WO 2007/053394 and Bioorg. Med. Chem. Lett. 2007, 17, 354-357).
  • a superior therapeutic index e.g. inhibitors that are at least equally efficacious and, in one or more respects, are less toxic at the relevant therapeutic dose than previous agents.
  • R 1 represents H or C 1-3 alkyl
  • R 2 and R 3 independently represent H or C 1-3 alkyl, or R 2 and R 3 together combine to form C 2-3 alkylene
  • X 1 and X 2 are both N, or X 1 is C and X 2 is either O or S
  • Ar is phenyl or a 5- or 6-membered heteroaryl group containing one or more heteroatoms selected from N, O and S, which phenyl and heteroaryl groups are optionally substituted by one or more substituents selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy, hydroxy, amino and cyano
  • L is a direct bond or C 1-2 alkylene
  • E represents:
  • salts include acid addition salts and base addition salts.
  • Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of formula I in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
  • Examples of pharmaceutically acceptable salts include acid addition salts derived from mineral acids and organic acids, and salts derived from metals.
  • compounds of formula I may contain the stated atoms in any of their natural or non-natural isotopic forms.
  • embodiments of the invention that may be mentioned include those in which:
  • references herein to an “isotopic derivative” relate to the second of these two embodiments.
  • the compound of formula I is isotopically enriched or labelled (with respect to one or more atoms of the compound) with one or more stable isotopes.
  • the compounds of the invention that may be mentioned include, for example, compounds of formula I that are isotopically enriched or labelled with one or more atoms such as deuterium or the like.
  • alkyl groups and alkoxy groups as defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of three) of carbon atoms, be branched.
  • Particular alkyl groups that may be mentioned include, for example, methyl, ethyl, n-propyl, iso-propyl, butyl, n-butyl and tert-butyl.
  • Particular alkoxy groups that may be mentioned include, for example, methoxy, ethoxy, propoxy, and butoxy.
  • cycloalkyl groups as defined herein may, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, be part cyclic/acyclic.
  • alkylene groups as defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be branched.
  • alkylene refers to straight-chain alkylene.
  • aryl groups may be via any atom of the ring system. However, when aryl groups are bicyclic or tricyclic, they are linked to the rest of the molecule via an aromatic ring.
  • C 6-14 aryl groups include phenyl, naphthyl and the like. Embodiments of the invention that may be mentioned include those in which aryl is phenyl.
  • Het a that may be mentioned include imidazolyl (e.g. imidazol-2-yl), isothiazolyl (e.g. isothiazol-3-yl), isoxazolyl (e.g. isoxazol-3-yl), 1,2,4-oxadiazolyl (e.g. 1,2,4-oxadiazol-3-yl or 1,2,4-oxadiazol-5-yl), 1,3,4-oxadiazolyl, oxazolyl (e.g. oxazol-2-yl), pyridinyl (e.g. pyridin-2-yl), pyrimidinyl (e.g.
  • 1,2,4-thiadiazolyl e.g. 1,2,4-thiadiazol-3-yl or 1,2,4-thiadiazol-5-yl
  • 1,3,4-thiadiazolyl thiazolyl (e.g. thiazol-2-yl)
  • 1,2,3-triazolyl e.g. 1,2,3-triazol-1-yl, 1,2,3-triazol-4-yl or 1,2,3-triazol-5-yl
  • 1,2,4-triazolyl e.g. 1,2,4-triazol-1-yl, 1,2,4-triazol-3-yl or 1,2,4-triazol-5-yl.
  • halo includes references to fluoro, chloro, bromo or iodo, in particular to fluoro, chloro or bromo, especially fluoro or chloro.
  • R 1 represents C 1-2 alkyl or, particularly, H; R 2 and R 3 independently represent C 1-2 alkyl, or R 2 and R 3 together combine to form C 2 alkylene; X 1 and X 2 are both N; Ar is pyrimidinyl or, particularly, phenyl or pyridinyl, which three groups are optionally substituted by one or more substituents selected from halo, C 1-3 alkyl and C 1-3 alkoxy; L is a direct bond; E represents H, halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl or C 1-6 alkoxy, which latter four groups are optionally substituted by one or more halo substituents or by NR 7a R 7b , R 7a and R 7b , independently on each occurrence, represent C 1-2 alkyl, or, together with the N-atom to which they are attached, form a 5- or 6-membered heterocyclic group that is fully saturated and which heterocyclic group contains one N
  • R 1 represents C 1-2 alkyl or, particularly, H; R 2 and R 3 independently represent H or C 1-2 alkyl, or R 2 and R 3 together combine to form C 2 alkylene;
  • A represents CH or N;
  • a 2 represents N or, particularly, CH;
  • R 4 and R 5 are both halo, or R 4 and R 5 , together with the C-atoms to which they are attached, form a fused phenyl ring;
  • R a represents C 1-2 alkoxy or, particularly, C 1-2 alkyl, which latter two groups are optionally substituted by one or more halo atoms, or R a represents C 1-2 alkyl or C 2 alkoxy, which latter two groups are substituted by NR 7a R 7b ;
  • R 7a and R 7b both represent C 1-2 alkyl or, together with the N-atom to which they are attached, form a 5- or 6-membered heterocyclic group that is fully saturated and which heterocyclic group contains one N atom (the atom
  • R b , R c and R d represents H, —C(O)NH—CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d or -Q 1 -CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d
  • the other two of R b , R c and R d represent H, halo, —C ⁇ C—H, C 1-4 alkyl or C 1-4 alkoxy, which latter two groups are optionally substituted by one or more halo atoms)
  • R b , R c and R d independently represent H, halo, hydroxy, C 1-4 alkylene-N(R 9c )R 9d , —C(O)NH—CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d , -Q 1 -CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d ,
  • R b , R c and R d represents H, —C(O)NH—CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d or -Q 1 -CH 2 —[C 1-3 alkylene]-N(R 9c )R 9d , and the other two of R b , R c and R d represent H, halo, C 1-4 alkyl or C 1-4 alkoxy, which latter two groups are optionally substituted by one or more halo atoms)), or R b and R c , together with the C-atoms to which they are attached, form a fused, 5- or 6-membered heteroaromatic or heterocyclic ring, which ring:
  • Still further embodiments of the invention include those in which the compound of formula Ia is a compound of formula Ib,
  • R 1 to R 5 , A, A 2 , A 3 , A 4 , R a to R d are as defined above in respect of compounds of formula Ia.
  • Embodiments of the invention that may be mentioned include those in which, in the compound of formula Ia or Ib:
  • R c and R d are as defined above in respect of compounds of formula Ia or Ib (such as wherein R c and R d are both H), and A x represents N or, particularly, CH.
  • R 1 to R 5 , A, A 2 , A x and R a are as defined above in respect of compounds of formula Ia and Ib;
  • R c2 and R d2 are defined above for R c and R d in respect of compounds of formula Ia and Ib;
  • R d1 is as defined above for R d in respect of compounds of formula Ia and Ib (e.g. R d1 is methyl).
  • Embodiments of the invention include those in which one or more of the following definitions apply to the compound of formula Ic, Id or Ie:
  • salts of compounds of formula I, Ia, Ib, Ic, Id or Ie include all pharmaceutically acceptable salts, such as, without limitation, acid addition salts of strong mineral acids such as HCl and HBr salts and addition salts of strong organic acids such as methanesulfonic acid.
  • references herein to a compound of the invention are intended to include references to the compound and to all pharmaceutically acceptable salts, solvates and/or tautomers of said compound, unless the context specifically indicates otherwise.
  • solvates that may be mentioned include hydrates.
  • the compounds of the invention are p38 MAP kinase inhibitors (especially of the alpha subtype) and are therefore useful in medicine, in particular for the treatment of inflammatory diseases. Further aspects of the invention that may be mentioned therefore include the following.
  • diluents and carriers that may be mentioned include those suitable for parenteral, oral, topical, mucosal and rectal administration.
  • compositions and combination products of aspects (a) and (b) above may be prepared e.g. for parenteral, subcutaneous, intramuscular, intravenous, intra-articular, intravitreous, periocular, retrobulbar, subconjunctival, sub-Tenon, topical ocular or peri-articular administration, particularly in the form of liquid solutions, emulsions or suspensions; for oral administration, particularly in the form of tablets or capsules, and especially involving technologies aimed at furnishing colon-targeted drug release (Patel, M. M. Expert Opin. Drug Deliv. 2011, 8 (10), 1247-1258); for topical e.g.
  • pulmonary or intranasal administration particularly in the form of powders, nasal drops or aerosols and transdermal administration; for topical ocular administration, particularly in the form of solutions, emulsions, suspensions, ointments, implants/inserts, gels, jellies or liposomal microparticle formulations (Ghate, D.; Edelhauser, H. F. Expert Opin. Drug Deliv. 2006, 3 (2), 275-287); for ocular administration, particularly in the form of biodegradable and non-biodegradable implants, liposomes and nanoparticles (Thrimawithana, T. R. et al. Drug Discov. Today 2011, 16 (5/6), 270-277); for mucosal administration e.g. to buccal, sublingual or vaginal mucosa, and for rectal administration e.g. in the form of a suppository or enema.
  • mucosal administration e.g. to buccal, sublingual or vagina
  • compositions and combination products of aspects (a) and (b) above may conveniently be administered in unit dosage form and may be prepared by any of the methods well-known in the pharmaceutical art, for example as described in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., (1985).
  • Formulations for parenteral administration may contain as excipients sterile water or saline, alkylene glycols such as propylene glycol, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes and the like.
  • Formulations for nasal administration may be solid and may contain excipients, for example, lactose or dextran, or may be aqueous or oily solutions for use in the form of nasal drops or metered sprays.
  • excipients include sugars, calcium stearate, magnesium stearate, pregelatinated starch, and the like.
  • Pharmaceutical formulations and combination products suitable for oral administration may comprise one or more physiologically compatible carriers and/or excipients and may be in solid or liquid form.
  • Tablets and capsules may be prepared with binding agents, for example, syrup, acacia, gelatin, sorbitol, tragacanth, or poly-vinylpyrollidone; fillers, such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, or glycine; lubricants, such as magnesium stearate, talc, polyethylene glycol, or silica; and surfactants, such as sodium lauryl sulfate.
  • binding agents for example, syrup, acacia, gelatin, sorbitol, tragacanth, or poly-vinylpyrollidone
  • fillers such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, or glycine
  • lubricants such as magnesium stearate, talc,
  • Liquid compositions may contain conventional additives such as suspending agents, for example sorbitol syrup, methyl cellulose, sugar syrup, gelatin, carboxymethyl-cellulose, or edible fats; emulsifying agents such as lecithin, or acacia; vegetable oils such as almond oil, coconut oil, cod liver oil, or peanut oil; preservatives such as butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT).
  • suspending agents for example sorbitol syrup, methyl cellulose, sugar syrup, gelatin, carboxymethyl-cellulose, or edible fats
  • emulsifying agents such as lecithin, or acacia
  • vegetable oils such as almond oil, coconut oil, cod liver oil, or peanut oil
  • preservatives such as butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT).
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluen
  • Solid oral dosage forms include tablets, two-piece hard shell capsules and soft elastic gelatin (SEG) capsules.
  • SEG soft elastic gelatin
  • a dry shell formulation typically comprises of about 40% to 60% w/w concentration of gelatin, about a 20% to 30% concentration of plasticizer (such as glycerin, sorbitol or propylene glycol) and about a 30% to 40% concentration of water. Other materials such as preservatives, dyes, opacifiers and flavours also may be present.
  • the liquid fill material comprises a solid drug that has been dissolved, solubilized or dispersed (with suspending agents such as beeswax, hydrogenated castor oil or polyethylene glycol 4000) or a liquid drug in vehicles or combinations of vehicles such as mineral oil, vegetable oils, triglycerides, glycols, polyols and surface-active agents.
  • a compound of the invention may be administered topically (e.g. to the lung, eye or intestines).
  • topically e.g. to the lung, eye or intestines.
  • embodiments of aspects (a) and (b) above that may be mentioned include pharmaceutical formulations and combination products that are adapted for topical administration.
  • Such formulations include those in which the excipients (including any adjuvant, diluent and/or carrier) are topically acceptable.
  • Aerosol formulations typically comprise the active ingredient suspended or dissolved in a suitable aerosol propellant, such as a chlorofluorocarbon (CFC) or a hydrofluorocarbon (HFC).
  • a suitable aerosol propellant such as a chlorofluorocarbon (CFC) or a hydrofluorocarbon (HFC).
  • CFC propellants include trichloromonofluoromethane (propellant 11), dichlorotetrafluoromethane (propellant 114), and dichlorodifluoromethane (propellant 12).
  • Suitable HFC propellants include tetrafluoroethane (HFC-134a) and heptafluoropropane (HFC-227).
  • the propellant typically comprises 40% to 99.5% e.g.
  • the formulation may comprise excipients including co-solvents (e.g. ethanol) and surfactants (e.g. lecithin, sorbitan trioleate and the like). Aerosol formulations are packaged in canisters and a suitable dose is delivered by means of a metering valve (e.g. as supplied by Bespak, Valois or 3M).
  • a metering valve e.g. as supplied by Bespak, Valois or 3M.
  • Topical administration to the lung may also be achieved by use of a non-pressurised formulation such as an aqueous solution or suspension. This may be administered by means of a nebuliser. Topical administration to the lung may also be achieved by use of a dry-powder formulation.
  • a dry powder formulation will contain the compound of the disclosure in finely divided form, typically with a mass mean aerodynamic diameter (MMAD) of 1-10 ⁇ m.
  • the formulation will typically contain a topically acceptable diluent such as lactose, usually of large particle size e.g. an MMAD of 100 ⁇ m or more. Examples of dry powder delivery systems include SPINHALER, DISKHALER, TURBOHALER, DISKUS and CLICKHALER.
  • the compounds of the present invention may also be administered rectally, for example in the form of suppositories or enemas, which include aqueous or oily solutions as well as suspensions and emulsions.
  • suppositories can be prepared by mixing the active ingredient with a conventional suppository base such as cocoa butter or other glycerides.
  • the drug is mixed with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • the total amount of the inhibitor will be about 0.0001 to less than 4.0% (w/w).
  • compositions administered according to the present invention will be formulated as solutions, suspensions, emulsions and other dosage forms.
  • Aqueous solutions are generally preferred, based on ease of formulation, as well as a patient's ability to administer such compositions easily by means of instilling one to two drops of the solutions in the affected eyes.
  • the compositions may also be suspensions, viscous or semi-viscous gels, or other types of solid or semi-solid compositions. Suspensions may be preferred for compounds that are sparingly soluble in water.
  • compositions administered according to the present invention may also include various other ingredients, including, but not limited to, tonicity agents, buffers, surfactants, stabilizing polymer, preservatives, co-solvents and viscosity building agents.
  • Preferred pharmaceutical compositions of the present invention include the inhibitor with a tonicity agent and a buffer.
  • the pharmaceutical compositions of the present invention may further optionally include a surfactant and/or a palliative agent and/or a stabilizing polymer.
  • tonicity agents may be employed to adjust the tonicity of the composition, preferably to that of natural tears for ophthalmic compositions.
  • sodium chloride, potassium chloride, magnesium chloride, calcium chloride, simple sugars such as dextrose, fructose, galactose, and/or simply polyols such as the sugar alcohols mannitol, sorbitol, xylitol, lactitol, isomaltitol, maltitol, and hydrogenated starch hydrolysates may be added to the composition to approximate physiological tonicity.
  • Such an amount of tonicity agent will vary, depending on the particular agent to be added.
  • compositions will have a tonicity agent in an amount sufficient to cause the final composition to have an ophthalmically acceptable osmolality (generally about 150-450 mOsm, preferably 250-350 mOsm and most preferably at approximately 290 mOsm).
  • ophthalmically acceptable osmolality generally about 150-450 mOsm, preferably 250-350 mOsm and most preferably at approximately 290 mOsm.
  • the tonicity agents of the invention will be present in the range of 2 to 4% w/w.
  • Preferred tonicity agents of the invention include the simple sugars or the sugar alcohols, such as D-mannitol.
  • An appropriate buffer system e.g., sodium phosphate, sodium acetate, sodium citrate, sodium borate or boric acid
  • the particular concentration will vary, depending on the agent employed.
  • the buffer will be chosen to maintain a target pH within the range of pH 5 to 8, and more preferably to a target pH of pH 5 to 7.
  • Surfactants may optionally be employed to deliver higher concentrations of inhibitor.
  • the surfactants function to solubilise the inhibitor and stabilise colloid dispersion, such as micellar solution, microemulsion, emulsion and suspension.
  • examples of surfactants which may optionally be used include polysorbate, poloxamer, polyosyl 40 stearate, polyoxyl castor oil, tyloxapol, triton, and sorbitan monolaurate.
  • Preferred surfactants to be employed in the invention have a hydrophile/lipophile/balance “HLB” in the range of 12.4 to 13.2 and are acceptable for ophthalmic use, such as TritonX114 and tyloxapol.
  • Additional agents that may be added to the ophthalmic compositions of the present invention are demulcents which function as a stabilising polymer.
  • the stabilizing polymer should be an ionic/charged example with precedence for topical ocular use, more specifically, a polymer that carries negative charge on its surface that can exhibit a zeta-potential of ( ⁇ )10-50 mV for physical stability and capable of making a dispersion in water (i.e. water soluble).
  • a preferred stabilising polymer of the invention would be polyelectrolyte, or polyelectrolytes if more than one, from the family of cross-linked polyacrylates, such as carbomers and Pemulen(R), specifically Carbomer 974p (polyacrylic acid), at 0.1-0.5% w/w.
  • viscosity enhancing agents include, but are not limited to: polysaccharides, such as hyaluronic acid and its salts, chondroitin sulfate and its salts, dextrans, various polymers of the cellulose family; vinyl polymers; and acrylic acid polymers.
  • Topical ophthalmic products are typically packaged in multidose form. Preservatives are thus required to prevent microbial contamination during use. Suitable preservatives include: benzalkonium chloride, chlorobutanol, benzododecinium bromide, methyl paraben, propyl paraben, phenylethyl alcohol, edentate disodium, sorbic acid, polyquaternium-1, or other agents known to those skilled in the art. Such preservatives are typically employed at a level of from 0.001 to 1.0% w/v. Unit dose compositions of the present invention will be sterile, but typically unpreserved. Such compositions, therefore, generally will not contain preservatives.
  • Embodiments of the invention that may be mentioned in connection with the combination products described at (b) above include those in which the other therapeutic agent is one or more therapeutic agents that are known by those skilled in the art to be suitable for treating inflammatory diseases (e.g. the specific diseases mentioned below).
  • the other therapeutic agent is one or more agents selected from the list comprising:
  • the other therapeutic agent may be, for example, one or more agents selected from the list comprising:
  • the other therapeutic agent may be, for example, one or more agents selected from the list comprising:
  • the compounds of the invention may be used as monotherapies for inflammatory diseases, or in combination therapies for such diseases.
  • embodiments of aspects (e) to (g) above include those in which the compound of formula I, Ia, Ib, Ic, Id or Ie (or pharmaceutically acceptable salt, solvate or isotopic derivative thereof) is the sole pharmacologically active ingredient utilised in the treatment.
  • the compound of formula I, la, Ib, Ic, Id or Ie (or pharmaceutically acceptable salt, solvate or isotopic derivative thereof) is administered to a subject who is also administered one or more other therapeutic agents (e.g. wherein the one or more other therapeutic agents are as defined above in connection with combination products).
  • inflammatory disease specifically includes references to any one or more of the following:
  • references herein to diseases having an inflammatory component include references to diseases that involve inflammation, whether or not there are other (non-inflammatory) symptoms or consequences of the disease.
  • E, L, Ar, X 1 , X 2 , R 1 to R 5 , A, A 1 , G and G 1 are as hereinbefore defined, for example under conditions known to those skilled in the art, for example at a temperature from ambient (e.g. 15 to 30° C.) to about 110° C. in the presence of a suitable organic solvent (e.g. a polar aprotic solvent such as DMF, THF, 1,4-dioxane, or mixtures thereof); (b) reaction of a compound of formula IIa,
  • Z 1 is as defined above, with a suitable azide-forming agent (i.e. a suitable source of a leaving group and activated azide ion, such as diphenyl phosphorazidate; see, for example, Tetrahedron 1974, 30, 2151-2157) under conditions known to those skilled in the art, such as at sub-ambient to ambient temperature (e.g. from an initial temperature of about ⁇ 5 to 5° C. to ambient temperature post-reaction) in the presence of an amine base (e.g. triethylamine or a sterically hindered base such as N,N-diisopropylethylamine) and a suitable organic solvent (e.g.
  • a suitable azide-forming agent i.e. a suitable source of a leaving group and activated azide ion, such as diphenyl phosphorazidate; see, for example, Tetrahedron 1974, 30, 2151-2157
  • a suitable azide-forming agent i.e. a suitable source of
  • reaction of a compound of formula IIb is followed, without isolation, by thermal rearrangement (e.g. under heating) of the intermediate acyl azide (of formula Z 1 —C(O)—N 3 ) e.g. at ambient temperature (such as from 15 to 30° C.) to provide, in situ, a compound of formula II, which compound is then reacted with a compound of formula III, as defined above, to provide the compound of formula I; (c) reaction of a compound of formula IIb,
  • LG 1 represents a suitable leaving group (e.g. imidazolyl, chloro, or aryloxy) and Z 1 is as defined above, with a compound of formula III, as defined above, for example under conditions known to those skilled in the art, such as at ambient temperature (e.g. from 15 to 30° C.), optionally in the presence of an amine base (e.g. a sterically hindered base like N,N-diisopropylethylamine) and a suitable organic solvent (e.g. an aprotic solvent, such as dichloromethane); (d) reaction of a compound of formula VI,
  • amine base e.g. a sterically hindered base like N,N-diisopropylethylamine
  • a suitable organic solvent e.g. an aprotic solvent, such as dichloromethane
  • LG 2 represents a suitable leaving group (e.g. a halo group such as chloro or bromo) and E, L, Ar, X 1 , X 2 , R 1 to R 5 , A and A 1 are as hereinbefore defined with a compound of formula VII,
  • G and G 1 are as hereinbefore defined, for example under conditions known to those skilled in the art (e.g. as described in J. Am. Chem. Soc. 2011, 133, 15686-15696), such as at elevated temperature (e.g. from 50 to 110° C.) in the presence of a suitable organic solvent (e.g. a polar aprotic solvent such as DMF, THF, 1,4-dioxane, or mixtures thereof) and, optionally, an acidic catalyst (e.g.
  • a suitable organic solvent e.g. a polar aprotic solvent such as DMF, THF, 1,4-dioxane, or mixtures thereof
  • an acidic catalyst e.g.
  • a sulfonic acid such as para-toluenesulfonic acid, for example in the presence of approximately 0.5 to 1 equivalents of such an acid relative to the compound of formula VI or formula VII); or (e) deprotection of an protected derivative of a compound of formula I, under conditions known to those skilled in the art, wherein the protected derivative bears a protecting group on an O- or N-atom of the compound of formula I (and, for the avoidance of doubt, a protected derivative of one compound of formula I may or may not represent another compound of formula I).
  • Compounds of formula II may be prepared according to or by analogy with methods known to those skilled in the art, for example by reaction of a compound of formula IIa, as defined above, with an azide-forming agent, followed by rearrangement of the intermediate acyl azide (as described at (b) above; see, for example, Tetrahedron 1974, 30, 2151-2157).
  • LG 1 is as hereinbefore defined, with a compound of formula IX,
  • Z 1 is as hereinbefore defined, for example under conditions known to those skilled in the art.
  • Amines of formula IX may be prepared from carboxylic acids of formula IIa through the route described in (b) above, where the intermediate isocyanate II is hydrolysed with water to give a carbamic acid that loses carbon dioxide to furnish IX.
  • the intermediate isocyanate II can be reacted with an alcohol, such as t-butanol, to generate a protected version of IX.
  • the remaining halogen or methanesulfonyl substituents (LG 2 ) of the ether XX is then displaced i) by an amine of formula VII in a second S N Ar reaction or (ii) via a Buchwald coupling (see, for example, WO 2009/017838) with an amine of formula VII to furnish the desired compound (when FG is NH 2 ), or XXI (when FG is nitro or NH-PG 2 ).
  • the NH 2 group may be revealed by a reduction reaction, typically done through hydrogenation employing a suitable catalyst, e.g., palladium on carbon, or employing dissolving metal conditions, such as with iron in glacial acetic acid.
  • the NH 2 group may be revealed by a deprotection reaction.
  • the unmasking of the latent NH 2 group represented by FG can take place at any stage in the synthetic route shown in Scheme 3.
  • Certain compounds of formula XIV may be synthesised employing the route highlighted in Scheme 4.
  • phenols XXIII are alkylated with alkyl halides XXIV, where m is 1-7 and Hal is chloro, bromo or iodo, under basic conditions in Williamson ether syntheses (see, for example: Eur. J. Med. Chem. 2010, 45, 5965-5978).
  • Phenols XXIII can be prepared from XXII—in which PG 1 is an appropriate protecting group that masks the reactive phenolic functionality (see, for example: Greene, T.
  • the compounds of formula XXII may be synthesised using, for example, the above-mentioned Chan-Lam reactions (Scheme 1) of compounds of formula XIII where L is a direct bond and E is O-PG 1 .
  • compounds of formula VI may be synthesised by analogy with the compounds of formula I (see, for example, alternative processes (a) to (c) above).
  • compounds of formula VI can be prepared by reaction of a compound of formula IIx with a compound of formula IIIx, wherein the compounds of formulae IIx and IIIx take the same definitions as the compounds of formulae II and III, with the exception that one of Z 1 and Z 2 represents a structural fragment of formula IV, as hereinbefore defined, and the other of Z 1 and Z 2 represents a structural fragment of formula Va,
  • Novel intermediates as described herein form an aspect of the invention.
  • further aspects of the invention relate to:
  • the groups E, L, Ar, X 1 , X 2 , R 1 to R 5 , A, A 1 and LG 2 when present, take any of the definitions for those groups as hereinbefore defined.
  • Protected derivatives of the compound of formula IIa include esters (e.g. C 1-4 alkyl esters) thereof.
  • Protected derivatives of the compound of formula III include those in which the essential NH 2 group is protected.
  • such protected derivatives include amides or, particularly, carbamates of those compounds.
  • those protected derivatives include compounds in which a H-atom of the NH 2 group is replaced by:
  • alkyne esters of formula XV may be commercially available or prepared by conventional methods, the present applicant has surprisingly discovered that the corresponding carboxylic acids of such esters may be advantageously prepared by a route involving carboxylation of a Grignard reagent containing a trialkylsilyl-protected alkyne.
  • R s1 to R s3 independently represent C 1-4 alkyl and R 2 and R 3 are as hereinbefore defined, said process comprising the steps of:
  • the compound of formula XXVI is prepared by reaction of the corresponding halo compound with magnesium metal.
  • step (i) of the above-described process may be replaced by the steps:
  • the compound of formula XXVI is formed in the same solvent system as used for the reaction with carbon dioxide (i.e. the compound of formula XXVII is reacted with carbon dioxide without isolation from the reaction mixture in which it is formed).
  • any one or more (e.g. all) of the following may apply.
  • R 2 and R 3 are as hereinbefore defined, said process comprising removal of the —Si(R s1 )(R s2 )(R s3 )protecting group from a compound of formula XXV, as hereinbefore defined, for example under conditions known to those skilled in the art (e.g. by contacting the compound of formula XXV at ambient temperature with a mixture of an alkali metal hydroxide (such as KOH) and a C 1-4 alkyl alcohol, such as methanol).
  • an alkali metal hydroxide such as KOH
  • a C 1-4 alkyl alcohol such as methanol
  • the compound of formula XXV is prepared according to a method as described above.
  • a particular embodiment of the invention relates to a process for the preparation of a compound of formula XXXI, as hereinbefore defined, said process comprising:
  • a process for the preparation of a compound of formula I comprising a process as hereinbefore defined for the preparation of a compound of formula XXV, XXXI or XV.
  • a further aspect of the invention relates to a compound of formula XXV, as hereinbefore defined, or a salt or protected derivative thereof.
  • Protected derivatives of the compound of formula XXV include esters (e.g. C 1-4 alkyl esters) thereof.
  • esters e.g. C 1-4 alkyl esters
  • Particular compounds of formula XXV that may be mentioned include those in which R s1 to R s3 , R 2 and R 3 take any of the definitions mentioned above for those groups (e.g. R s1 to R s3 , R 2 and R 3 all represent methyl).
  • the other aspects of the invention described herein may have the advantage that, in the treatment of the conditions described herein, they may be more convenient for the physician and/or patient than, be more efficacious than, be less toxic than, have better selectivity over, have a broader range of activity than, be more potent than, produce fewer side effects than, have a better pharmacokinetic and/or pharmacodynamic profile than, have more suitable solid state morphology than, have better stability (e.g. long term stability) than, or may have other useful pharmacological properties over, similar compounds, combinations, methods (treatments) or uses known in the prior art for use in the treatment of those conditions or otherwise.
  • the compounds of the invention may additionally (or alternatively):
  • Analytical HPLC was carried out using an Agilent Zorbax Extend C18, Rapid Resolution HT 1.8 ⁇ m column eluting with a gradient of either 0.1% formic acid in MeCN in 0.1% aqueous formic acid or a gradient of MeCN in 10 mM Ammonium Bicarbonate; a Waters Xselect CSH C18 3.5 ⁇ m eluting with a gradient of 0.1% formic acid in MeCN in 0.1% aqueous formic acid. UV spectra of the eluted peaks were measured using either a diode array or variable wavelength detector on an Agilent 1100 system.
  • Analytical LCMS was carried out using an Agilent Zorbax Extend C18, Rapid Resolution HT 1.8 ⁇ m column eluting with a gradient of either 0.1% formic acid in MeCN in 0.1% aqueous formic acid or a gradient of MeCN in 10 mM Ammonium Bicarbonate; a Waters Xselect CSH C18 3.5 ⁇ m eluting with a gradient of 0.1% formic acid in MeCN in 0.1% aqueous formic acid.
  • UV and mass spectra of the eluted peaks were measured using a variable wavelength detector on either an Agilent 1100 with or an Agilent Infinity 1260 LC with 6120 quadrupole mass spectrometer with positive and negative ion electrospray.
  • Preparative HPLC was carried out using an Agilent Prep-C18 5 ⁇ m Preparative Cartridge using either a gradient of either 0.1% formic acid in MeCN in 0.1% aqueous formic acid or a gradient of MeCN in 10 mM Ammonium Bicarbonate; or a Waters Xselect CSH C18 5 ⁇ m column using a gradient 0.1% MeCN in 0.1% aqueous formic acid. Fractions were collected following detection by UV at 254 nm.
  • Pre-ground (mortar and pestle) magnesium turnings (4.1 g, 0.172 mol) were charged to a 3-neck flask containing THF (75 mL). The mixture was stirred under N 2 at room temperature whilst ⁇ 3 g of (3-chloro-3-methylbut-1-yn-1-yl)trimethylsilane (see step (ii) above) was added. Once initiation of reaction was observed (exotherm!) the remaining (3-chloro-3-methylbut-1-yn-1-yl)trimethylsilane (12 g; reaction total 15 g, 0.086 mol) was added with cooling so as to keep the temperature in the range 15-22° C.
  • reaction mixture was stirred at room temperature for a further 1 hour before being added to solid CO 2 (150 g). After 1 hour, when there was no longer any solid CO 2 present, water was added and the mixture made acidic with HCl. The mixture was extracted with Et 2 O, the organic extracts dried (MgSO 4 ) and concentrated under reduced pressure to give 2,2-dimethyl-4-(trimethylsilyl)but-3-ynoic acid (15.86 g, 100%) as a yellow oil that solidified on cooling.
  • p-TSA monohydrate (2.80 g, 14.72 mmol) was added to a stirred mixture of 4-((2-chloropyrimidin-4-yl)oxy)naphthalen-1-amine (see, for example, Cirillo, P. F. et al., WO 2002/92576, 21 Nov. 2000; 8 g, 29.4 mmol) and aniline (6.71 mL, 73.6 mmol) in THF (50 mL) at rt under N 2 . The mixture was heated under reflux for 2 h (reaction mixture solidified), a further 50 ml of THF was added and the mixture heated for a further 2 h.
  • the aqueous layer was acidified with 1 M hydrogen chloride (20 mL) and extracted with diethyl ether (3 ⁇ 25 mL). The combined organic phases were washed with saturated brine (25 mL) and dried (MgSO 4 ). The solvent was removed under reduced pressure to yield the sub-title compound (950 mg).
  • the pre-adsorbed silica was purified by chromatography on the Companion (40 g column, 0-20% ethyl acetate:hexanes) to afford the sub-title compound (533 mg) as a dark brown oil which crystallised on standing.
  • step (iii) above 134 mg, 0.560 mmol
  • CDI 100 mg, 0.616 mmol
  • the product from step (i) above 184 mg, 0.560 mmol) in THF (2 mL) and stirring at rt for 4 h.
  • the mixture was concentrated under reduced pressure and the residue was triturated in MeCN to yield 180 mg of cream solid.
  • the solid was purified by chromatography on silica gel (12 g column, 10-50% ethyl acetate/isohexane to afford the product as a cream solid.
  • the solid was co-evaporated with DCM, triturated in methanol then recrystallised in acetonitrile to yield the title compound (96 mg) as a white solid.
  • Phenyl chloroformate (2.65 ml, 21.06 mmol) was added to a stirred mixture of 4-((2-chloropyrimidin-4-yl)oxy)naphthalen-1-amine (see, for example, Cirillo, P. F. et al., WO 2002/92576, 21 Nov. 2000; 5.45 g, 20.06 mmol) and sodium bicarbonate (3.37 g, 40.1 mmol) in DCM (50 ml) and THF (20 ml) at rt. The mixture was stirred for 18 h then a further portion of phenyl chloroformate added (0.5 mL) and left for 2 h.
  • the crude product was purified by chromatography on the Companion (40 g column, 0-5% MeOH/1M NH 3 in DCM) to afford an orange solid which was repurified by chromatography on silica gel (25 g column, 0-50% acetone in toluene) then by prep HPLC (Waters, Acidic (0.1% Formic acid), Waters X-Select Prep-C18, 5 ⁇ m, 19 ⁇ 50 mm column, 40-70% MeCN in Water) to afford the product as partial formate salts.
  • the product was loaded onto a column of SCX (2 g) in MeOH (2 mL).
  • Phenyl chloroformate 38 ⁇ L, 0.302 mmol was added to a stirred mixture of sodium bicarbonate (46.2 mg, 0.550 mmol) and the product from step (i) above (90 mg, 0.275 mmol) in THF (0.6 mL) and DCM (1.5 mL) and the reaction stirred for 2 days at ambient temperature. After this time the reaction was partitioned between DCM (30 mL) and brine (20 mL), the organic phase separated, dried (MgSO 4 ) and evaporated under reduced pressure.
  • the DCM layer was isolated by passing the mixture through a phase separation cartridge and the solvent evaporated and the crude material purified by preparative HPLC (Waters, Acidic (0.1% Formic acid), Waters X-Select Prep-C18, 5 ⁇ m, 19 ⁇ 50 mm column, 30-70% MeCN in Water) to afford the title compound (0.3Formic Acid present) (23 mg) as a white powder.
  • preparative HPLC Waters, Acidic (0.1% Formic acid), Waters X-Select Prep-C18, 5 ⁇ m, 19 ⁇ 50 mm column, 30-70% MeCN in Water
  • the sub-title compound can be prepared according to or by analogy with procedures known to those skilled in the art and/or described herein. For example, the following procedure can be used.
  • step (ii) above To the product from step (ii) above (787 mg, 2.19 mmol) and the product of step (i) above (636 mg, 2.190 mmol) in isopropyl acetate (15 mL) was added Et 3 N (45.8 ⁇ L, 0.329 mmol) and the reaction heated at 60° C. over 16 h. The reaction was allowed to cool to ambient temperature and a white precipitate was removed by filtration. Filtrate was concentrated under reduced pressure and purified by chromatography on silica gel (40 g column, 2% Ethanol in toluene) to afford the sub-title compound (200 mg) as a pale brown powder (approx 65% purity).
  • the crude product was purified by chromatography on the Companion (40 g column, 20-50% ethyl acetate in toluene) to afford a yellow powder (44 mg).
  • the crude product was purified by preparative HPLC (Waters, Acidic (0.1% Formic acid), Waters X-Select Prep-C18, 5 ⁇ m, 19 ⁇ 50 mm column, 35-75% MeCN in Water) to afford the title compound (20 mg) as a white powder.
  • 2-(Dimethylamino)ethanol (3.0 ml, 29.8 mmol) was treated with sodium (0.114 g, 4.98 mmol) in a sealed tube and stirred at ambient temperature until homogenous.
  • 6-Chloropyridin-2-amine (1.1 g, 8.30 mmol) was added and the reaction mixture was heated to 145° C. and stirred in a microwave tube for 4 h.
  • the reaction was cooled to rt then partitioned between EtOAc (50 mL) and water (30 mL). The aqueous phase was extracted with EtOAc (2 ⁇ 50 mL).
  • the reaction was cooled to rt.
  • the crude product was loaded directly onto a column of SCX.
  • the column was washed with MeOH and then the product was eluted with 1% NH 3 in MeOH.
  • the product-containing fraction was concentrated in vacuo to afford crude product as a dark brown semi-solid (1.85 g).
  • the crude product was dissolved in DCM (30 mL), TFA (10 mL, 130 mmol) was added and the reaction mixture stirred at rt for 3 h. The solvent was removed in vacuo and the resulting dark, brown residue dissolved in the minimum quantity of MeOH and loaded onto SCX.
  • the column was eluted with MeOH followed by 1% NH 3 in MeOH.
  • the reaction was cooled to rt.
  • the crude product was loaded directly onto a column of SCX in MeOH-DCM.
  • the column was washed with MeOH and then the product was eluted with 1% NH 3 in MeOH.
  • the product-containing fraction was concentrated in vacuo to afford crude product as a dark brown oil (1.66 g).
  • the crude product was dissolved in DCM (25 mL).
  • TFA (8 mL, 104 mmol) was added and the reaction mixture stirred at rt for 3 h. The solvent was removed in vacuo and the resulting dark, brown residue dissolved in the minimum quantity of MeOH and loaded onto SCX.
  • the column was eluted with MeOH followed by 1% NH 3 in MeOH.
  • Triethylamine (7.19 ⁇ l, 0.052 mmol) was added to a stirred mixture phenyl (3-(2-methylbut-3-yn-2-yl)-1-(p-tolyl)-1H-pyrazol-5-yl)carbamate (see Example 9(ii) above; 159 mg, 0.333 mmol) and the product from step (ii) above (169 mg, 0.333 mmol).
  • the resulting mixture was heated at 70° C. for 30 min.
  • the reaction was cooled to rt then partitioned between EtOAc (40 mL) and brine (20 mL).
  • Triethylamine (4 ⁇ L, 0.029 mmol) was added to a mixture of phenyl (3-(2-methylbut-3-yn-2-yl)-1-(p-tolyl)-1H-pyrazol-5-yl)carbamate (see Example 9(ii) above; 50 mg, 0.139 mmol) and the product of step (iii) above (73 mg, 0.140 mmol) in isopropyl acetate (2 mL) and the mixture heated at 50° C. for 4 h.
  • the crude product was loaded onto a column of SAX (Discovery® DSC-SAX, a polymer-bound quaternary amine) in MeOH.
  • SAX Discovery® DSC-SAX, a polymer-bound quaternary amine
  • the column was washed with MeOH and then the product was eluted with 5% AcOH in MeOH.
  • the resultant mixture was concentrated in vacuo then loaded onto a column of SCX in MeOH.
  • the column was washed with MeOH and then the product was eluted with 0.7 M ammonia in MeOH.
  • the resultant mixture was concentrated in vacuo to afford the sub-title compound (450 mg) as a yellow powder.
  • Pd(PPh 3 ) 4 (2.90 g, 2.51 mmol) was added to a degassed suspension of the product of step (i) above (16.5 g, 50.3 mmol), Cul (0.479 g, 2.51 mmol), and ethynyltriisopropyl-silane (16.92 mL, 75 mmol) in TEA (30 mL) and DMF (150 mL). Heated at 85° C. (block temp.) for 5 h, cooled and filtered (Whatman glass fibre pad GF/C). Solvents evaporated and the residue partitioned between EtOAc (500 mL) and 20% w/w NaCl soln. (500 mL).
  • step (ii) above (18.5 g, 43.1 mmol) was dissolved in EtOAc (250 mL) and TBAF, 1M in THF (43.1 mL, 43.1 mmol) added. Stirred for 1 h. Partitioned between water (500 mL) and ethyl acetate (200 mL) organic layer separated and washed with 20% w/w NaCl soln. (400 mL). Organic layer was separated, dried (MgSO 4 ) filtered and solvents evaporated. The crude product was slurried in Et 2 O (100 mL) for 30 minutes filtered and washed with fresh Et 2 O (20 mL). Oven dried at 45° C. to afford the sub-title compound (9.2 g)
  • the crude product was purified by chromatography on silica gel (40 g column, 0-10% MeOH in DCM) to afford a semi-solid, which was triturated with a mixture of diethyl ether, acetonitrile and isohexane to afford the title compound (165 mg) as a white solid.
  • the resultant mixture was concentrated in vacuo to afford the product as a dark brown oil.
  • the crude product was purified by chromatography on silica gel (80 g column, DCM:MeOH:NH 3 ; 95:5:0.05) to afford the sub-title compound (1.19 g) as a dark brown gum.
  • step (i) above To a microwave vial was added the product of step (i) above (379 mg, 1.479 mmol), tert-butyl (4-((2-chloropyrimidin-4-yl)oxy)naphthalen-1-yl)carbamate (see, for example, Ito, K. et al., WO 2010/067130, 17 Jun. 2010; 500 mg, 1.345 mmol), caesium carbonate (657 mg, 2.017 mmol), palladium(II) acetate (7.55 mg, 0.034 mmol), BINAP (41.9 mg, 0.067 mmol) and dioxane (8 mL). The reaction mixture was purged with N 2 for 10 min and then irradiated at 100° C.
  • the crude product was purified by chromatography on silica gel (80 g column, 0-10% MeOH in DMC) then purified further by chromatography on silica gel (80 g column, iso-hexane:ethyl acetate 1:1). Triturated with ether (2 ⁇ 2 mL) and the resultant solid was filtered, rinsing with ether, and dried in vacuo to afford the title compound (34 mg) as a clear white crystalline solid.
  • the enzyme inhibitory activities of compounds disclosed herein are determined by FRET using synthetic peptides labelled with both donor and acceptor fluorophores (Z-LYTE, Invitrogen Ltd., Paisley, UK).
  • the following two assay variants can be used for determination of p38 MAPK ⁇ inhibition.
  • the inhibitory activities of test compounds against the p38 MAPK ⁇ isoform are evaluated indirectly by determining the level of activation/phosphorylation of the down-stream molecule, MAPKAP-K2.
  • the p38 MAPK ⁇ protein (80 ng/mL, 2.5 ⁇ L) is mixed with the test compound (2.5 ⁇ L of either 4 ⁇ g/mL, 0.4 ⁇ g/mL, 0.04 ⁇ g/mL or 0.004 ⁇ g/mL) for 2 hr at RT.
  • the mix solution (2.5 ⁇ L) of the p38a inactive target MAPKAP-K2 (Invitrogen, 600 ng/mL) and FRET peptide (8 ⁇ M; a phosphorylation target for MAPKAP-K2) is then added and the kinase reaction is initiated by adding ATP (40 ⁇ M, 2.5 ⁇ L).
  • the mixture is incubated for 1 hr at RT.
  • Development reagent prote, 5 ⁇ L
  • This method follows the same steps as Method 1 above, but utilises a higher concentration of the p38 MAPK ⁇ protein (2.5 ⁇ L of 200 ng/mL protein instead of 2.5 ⁇ L of 80 ng/mL protein) for mixing with the test compound.
  • the inhibitory activities of compounds of the invention against p38MAPK ⁇ are evaluated in a similar fashion to that described hereinabove.
  • the enzyme (800 ng/mL, 2.5 ⁇ L) is incubated with the test compound (2.5 ⁇ L at either 4 ⁇ g/mL, 0.4 ⁇ g/mL, 0.04 ⁇ g/mL, or 0.004 ⁇ g/mL) for 2 hr at RT.
  • the FRET peptides (8 ⁇ M, 2.5 ⁇ L), and appropriate ATP solution (2.5 ⁇ L, 400 ⁇ M) is then added to the enzymes/compound mixtures and incubated for 1 hr.
  • Development reagent (protease, 5 ⁇ L) is added for 1 hr prior to detection in a fluorescence microplate reader (Varioskan® Flash, Thermo Scientific).
  • the inhibitory activities of compounds of the invention against c-Src and Syk enzymes are evaluated in a similar fashion to that described hereinabove.
  • the relevant enzyme (3000 ng/mL or 2000 ng/mL respectively, 2.5 ⁇ L) is incubated with the test compound (either 4 ⁇ g/mL, 0.4 ⁇ g/mL, 0.04 ⁇ g/mL, or 0.004 ⁇ g/mL, 2.5 ⁇ L each) for 2 hr at RT.
  • the FRET peptides (8 ⁇ M, 2.5 ⁇ L), and appropriate ATP solutions (2.5 ⁇ L, 800 ⁇ M for c-Src, and 60 ⁇ M ATP for Syk) are then added to the enzymes/compound mixtures and incubated for 1 hr. Development reagent (protease, 5 ⁇ L) is added for 1 hr prior to detection in a fluorescence microplate reader (Varioskan® Flash, ThermoFisher Scientific).
  • the following two assay variants can be used for determination of GSK 3 ⁇ inhibition.
  • the inhibitory activities of compounds of the invention against the GSK 3 ⁇ enzyme isoform are evaluated by determining the level of activation/phosphorylation of the target peptide.
  • the GSK3- ⁇ protein 500 ng/mL, 2.5 ⁇ L
  • the test compound 2.5 ⁇ L at either 4 ⁇ g/mL, 0.4 ⁇ g/mL, 0.04 ⁇ g/mL, or 0.004 ⁇ g/mL for 2 hr at RT.
  • the FRET peptide (8 ⁇ M, 2.5 ⁇ L), which is a phosphorylation target for GSK3 ⁇ , and ATP (40 ⁇ M, 2.5 ⁇ L) are then added to the enzyme/compound mixture and the resulting mixture incubated for 1 hr. Development reagent (protease, 5 ⁇ L) is added for 1 hr prior to detection in a fluorescence microplate reader (Varioskan® Flash, ThermoFisher Scientific).
  • the site-specific protease cleaves non-phosphorylated peptide only and eliminates the FRET signal.
  • Phosphorylation levels of each reaction are calculated using the ratio of coumarin emission (donor) over fluorescein emission (acceptor), for which high ratios indicate high phosphorylation and low ratios indicate low phosphorylation levels.
  • the percentage inhibition of each reaction is calculated relative to non-inhibited control and the 50% inhibitory concentration (IC 50 value) is then calculated from the concentration-response curve.
  • This method follows the same steps as Method 1 above, but utilises a shorter period of mixing of the test compound (105 minutes instead of 2 hours) with the GSK3- ⁇ protein.
  • the compounds of the invention were studied using one or more of the following assays.
  • U937 cells a human monocytic cell line
  • macrophage-type cells by incubation with phorbol myristate acetate (PMA; 100 ng/mL) for 48 to 72 hr.
  • PMA phorbol myristate acetate
  • Cells are pre-incubated with final concentrations of test compound for 2 hr and are then stimulated with 0.1 ⁇ g/mL of LPS (from E. Coli : O111:B4, Sigma) for 4 hr.
  • LPS from E. Coli : O111:B4, Sigma
  • the supernatant is collected for determination of TNF ⁇ and IL-8 concentrations by sandwich ELISA (Duo-set, R&D systems).
  • the inhibition of TNF ⁇ production is calculated as a percentage of that achieved by 10 ⁇ g/mL of BIRB796 at each concentration of test compound by comparison against vehicle control.
  • the relative 50% effective concentration (REC 50 ) is determined from the resultant concentration-response curve.
  • the inhibition of IL-8 production is calculated at each concentration of test compound by comparison with vehicle control.
  • the 50% inhibitory concentration (IC 50 ) is determined from the resultant concentration-response curve.
  • PBMCs Peripheral blood mononuclear cells
  • PBMCs Peripheral blood mononuclear cells
  • the PBMCs are seeded in 96 well plates and treated with compounds at the desired concentration for 2 hours before addition of 1 ng/mL LPS ( Escherichia Coli 0111:B4 from Sigma Aldrich) for 24 hours under normal tissue culture conditions (37° C., 5% CO 2 ).
  • LPS Escherichia Coli 0111:B4 from Sigma Aldrich
  • the supernatant is harvested for determination of IL-8 and TNF ⁇ concentrations by sandwich ELISA (Duo-set, R&D systems) and read on the fluorescence microplate reader (Varioskan® Flash, ThermoFisher Scientific).
  • the concentration at 50% inhibition (IC 50 ) of IL-8 and TNF ⁇ production is calculated from the dose response curve.
  • PBMCs from healthy subjects are separated from whole blood using a density gradient (Lymphoprep, Axis-Shield Healthcare). Cells are added to a 96 well plate pre-coated with a mixture of CD3/CD38 monoclonal antibodies (0.3 ⁇ g/mL eBioscience and 3 ⁇ g/mL BD Pharmingen respectively). Compound at the desired concentration is then added to the wells and the plate left for 3 days under normal tissue culture conditions. Supernatants are harvested and IL-2 and IFN gamma release determined by Sandwich ELISA (Duo-set, R&D System). The IC 50 is determined from the dose response curve.
  • HT29 cells a human colon adenocarcinoma cell line
  • IL-1 ⁇ IL-1 ⁇
  • supernatants are harvested for IL-8 quantification by Sandwich ELISA (Duo-set, R&D System). The IC 50 is determined from the dose response curve.
  • PBMCs from healthy subjects are separated from whole blood using a density gradient (Lymphoprep, Axis-Shield Healthcare). Cells are incubated for 2 hrs and non-adherent cells removed by washing. To differentiate the cells to macrophages the cells are incubated with 5 ng/mL of GM-CSF (Peprotech) for 7 days under normal tissue culture conditions. Compounds are then added to the cells at the desired concentration for a 2 hour pre-treatment before stimulation with 10 ng/mL LPS for 24 hours. Supernatants are harvested and IL-8 and TNF ⁇ release determined by Sandwich ELISA (Duo-set, R&D System). The IC 50 is determined from the dose response curve.
  • Poly I:C is used in these studies as a simple, RNA virus mimic.
  • Poly I:C-Oligofectamine mixture (1 ⁇ g/mL Poly I:C, ⁇ 2% Oligofectamine, 25 ⁇ L; Invivogen Ltd., San Diego, Calif., and Invitrogen, Carlsbad, Calif., respectively) is transfected into BEAS2B cells (human bronchial epithelial cells, ATCC). Cells are pre-incubated with final concentrations of test compounds for 2 hr and the level of ICAM1 expression on the cell surface is determined by cell-based ELISA.
  • the cells are washed with PBS-Tween (3 ⁇ 200 ⁇ L) and incubated with the secondary antibody (100 ⁇ L; HRP-conjugated anti-rabbit IgG, Dako Ltd., Glostrup, Denmark). The cells are then incubated with of substrate (50 ⁇ L) for 2-20 min, followed by the addition of stop solution (50 ⁇ L, 1N H 2 SO 4 ).
  • the ICAM-1 signal is detected by reading the absorbance at 450 nm against a reference wavelength of 655 nm using a spectrophotometer.
  • the cells are then washed with PBS-Tween (3 ⁇ 200 ⁇ L) and total cell numbers in each well are determined by reading absorbance at 595 nm after Crystal Violet staining (50 ⁇ L of a 2% solution in PBS) and elution by 1% SDS solution (100 ⁇ L) in distilled water.
  • the measured OD 450-655 readings are corrected for cell number by dividing with the OD595 reading in each well.
  • the inhibition of ICAM-1 expression is calculated at each concentration of test compound by comparison with vehicle control.
  • the 50% inhibitory concentration (IC 50 ) is determined from the resultant concentration-response curve.
  • PBMCs Peripheral blood mononucleocytes from healthy subjects are separated from whole blood (Quintiles, London, UK) using a density gradient (Histopaque®-1077, Sigma-Aldrich, Poole, UK).
  • the PBMCs (3 million cells per sample) are subsequently treated with 2% PHA (phytohaemagglutinin, Sigma-Aldrich, Poole, UK) for 48 hr, followed by a 20 hr exposure to varying concentrations of test compounds.
  • PHA phytohaemagglutinin, Sigma-Aldrich, Poole, UK
  • demecolcine 0.1 ⁇ g/mL; Invitrogen, Paisley, UK
  • PBMCs are permeabilised and fixed by adding Intraprep (50 ⁇ L; Beckman Coulter, France), and stained with anti-phospho-histone 3 (0.26 ng/L; #9701; Cell Signalling, Danvers, Mass.) and propidium iodide (1 mg/mL; Sigma-Aldrich, Poole, UK) as previously described (Muehlbauer P. A. and Schuler M. J., Mutation Research, 2003, 537:117-130). Fluorescence is observed using an ATTUNE flow cytometer (Invitrogen, Paisley, UK), gating for lymphocytes. The percentage inhibition of mitosis is calculated for each treatment relative to vehicle (0.5% DMSO) treatment.
  • Human rhinovirus RV16 is obtained from the American Type Culture Collection (Manassas, Va.). Viral stocks are generated by infecting Hela cells with HRV until 80% of the cells are cytopathic.
  • BEAS2B cells are infected with HRV at an MOI of 5 and incubated for 2 hr at 33° C. with gentle shaking for to promote absorption. The cells are then washed with PBS, fresh media added and the cells are incubated for a further 72 hr. The supernatant is collected for assay of IL-8 concentrations using a Duoset ELISA development kit (R&D systems, Minneapolis, Minn.).
  • the level of ICAM1 expressing cell surface is determined by cell-based ELISA.
  • cells are fixed with 4% formaldehyde in PBS. After quenching endogenous peroxidase by adding 0.1% sodium azide and 1% hydrogen peroxide, wells are washed with wash-buffer (0.05% Tween in PBS: PBS-Tween). After blocking well with 5% milk in PBS-Tween for 1 hr, the cells are incubated with anti-human ICAM-1 antibody in 5% BSA PBS-Tween (1:500) overnight. Wells are washed with PBS-Tween and incubated with the secondary antibody (HRP-conjugated anti-rabbit IgG, Dako Ltd.).
  • the ICAM-1 signal is detected by adding substrate and reading at 450 nm with a reference wavelength of 655 nm using a spectrophotometer.
  • the wells are then washed with PBS-Tween and total cell numbers in each well are determined by reading absorbance at 595 nm after Crystal Violet staining and elution by 1% SDS solution.
  • the measured OD 450-655 readings are corrected for cell number by dividing with the OD 595 reading in each well.
  • Compounds are added 2 hr before HRV infection and 2 hr after infection when non-infected HRV is washed out.
  • MRC-5 cells are infected with HRV16 at an MOI of 1 in DMEM containing 5% FCS and 1.5 mM MgCl 2 , followed by incubation for 1 hr at 33° C. to promote adsorption. The supernatants are aspirated, and then fresh media added followed by incubation for 4 days. Where appropriate, cells are pre-incubated with compound or DMSO for 2 hr, and the compounds and DMSO added again after washout of the virus.
  • Supernatants are aspirated and incubated with methylene blue solution (100 ⁇ L, 2% formaldehyde, 10% methanol and 0.175% Methylene Blue) for 2 hr at RT. After washing, 1% SDS in distilled water (100 ⁇ L) is added to each well, and the plates are shaken lightly for 1-2 hr prior to reading the absorbance at 660 nm. The percentage inhibition for each well is calculated. The IC 50 value is calculated from the concentration-response curve generated by the serial dilutions of the test compounds.
  • Normal human bronchial epithelial cells grown in 96 well plates are infected with RSV A2 (Strain A2, HPA, Salisbury, UK) at an MOI of 0.001 in the LHC8 Media:RPMI-1640 (50:50) containing 15 mM magnesium chloride and incubated for 1 hr at 37° C. for adsorption. The cells are then washed with PBS (3 ⁇ 200 ⁇ L), fresh media (200 ⁇ L) is added and incubation continued for 4 days. Where appropriate, cells are pre-incubated with the compound or DMSO for 2 hr, and then added again after washout of the virus.
  • the cells are fixed with 4% formaldehyde in PBS solution (50 ⁇ L) for 20 min, washed with WB (3 ⁇ 200 ⁇ L), (washing buffer, PBS including 0.5% BSA and 0.05% Tween-20) and incubated with blocking solution (5% condensed milk in PBS) for 1 hr. Cells are then washed with WB (3 ⁇ 200 ⁇ L) and incubated for 1 hr at RT with anti-RSV (2F7) F-fusion protein antibody (40 ⁇ L; mouse monoclonal, lot 798760, Cat. No.ab43812, Abcam) in 5% BSA in PBS-tween.
  • Differentiated U937 cells are pre-incubated with each test compound (final concentration 1 ⁇ g/mL or 10 ⁇ g/mL in 200 ⁇ L media indicated below) under two protocols: the first for 4 hr in 5% FCS RPMI1640 media and the second in 10% FCS RPMI1640 media for 24 h.
  • the supernatant is replaced with new media (200 ⁇ L) and MTT stock solution (10 ⁇ L, 5 mg/mL) is added to each well.
  • MTT stock solution 10 ⁇ L, 5 mg/mL
  • DMSO 200 ⁇ L
  • the percentage loss of cell viability is calculated for each well relative to vehicle (0.5% DMSO) treatment. Consequently an apparent increase in cell viability for drug treatment relative to vehicle is tabulated as a negative percentage.
  • Intestinal mucosa biopsies are obtained from the inflamed regions of the colon of IBD patients.
  • the biopsy material is cut into small pieces (2-3 mm) and placed on steel grids in an organ culture chamber at 37° C. in a 5% CO 2 /95% O 2 atmosphere in serum-free media.
  • DMSO control or test compounds at the desired concentration are added to the tissue and incubated for 24 hr in the organ culture chamber.
  • the supernatant is harvested for determination of IL-6, IL-8, IL-1 ⁇ and TNF ⁇ levels by R&D ELISA. Percentage inhibition of cytokine release by the test compounds is calculated relative to the cytokine release determined for the DMSO control (100%).
  • U937 cells a human monocytic cell line
  • PCA a human monocytic cell line
  • the cells are then incubated with either final concentrations of test compound or vehicle for 18 hr.
  • the induction of ⁇ -catenin by the test compounds is stopped by replacing the media with 4% formaldehyde solution.
  • Endogenous peroxide activity is neutralised by incubating with quenching buffer (100 ⁇ L, 0.1% sodium azide, 1% H 2 O 2 in PBS with 0.05% Tween-20) for 20 min.
  • quenching buffer 100 ⁇ L, 0.1% sodium azide, 1% H 2 O 2 in PBS with 0.05% Tween-20
  • the cells are washed with washing buffer (200 ⁇ L; PBS containing 0.05% Tween-20) and incubated with blocking solution (200 ⁇ L; 5% milk in PBS) for 1 hr, re-washed with washing buffer (200 ⁇ L) and then incubated overnight with anti- ⁇ -catenin antibody solution (50 ⁇ L) in 1% BSA/PBS (BD, Oxford, UK).
  • the measured OD 450-655 readings are corrected for cell number by dividing the OD 450-655 by the OD 595 readings.
  • the percentage induction for each well is calculated relative to vehicle, and the ratio of induction normalised in comparison with the induction produced by a standard control comprising of N-(4-(4-(3-(3-tert-butyl-1-p-tolyl-1H-pyrazol-5-yl)ureido)naphthalen-1-yloxy)pyridin-2-yl)-2-methoxyacetamide (1 ⁇ g/mL) which is defined as unity.
  • a signal less than 0.15 of that observed for the standard control is designated as “-ve”.
  • PBMCs from healthy subjects are separated from whole blood using a density gradient (Lymphoprep, Axis-Shield Healthcare).
  • the lymphocyte fraction is first enriched for CD4+ T cells by negative magnetic cell sorting as per the manufacturer's instructions (Miltenyi Biotec 130-091-155). Na ⁇ ve CD4+ T cells are then separated using positive magnetic selection of CD45RA+ cells using microbeads as per the manufacturer's instructions (130-045-901).
  • Cells are plated at 2 ⁇ 10 5 cells per well in 100 ⁇ L RPMI/10% FBS on 96 well flat bottomed plate (Corning Costar).
  • test compound 25 ⁇ L of test compound are diluted to the appropriate concentration (8 ⁇ final conc.) in normal medium and added to duplicate wells on the plate to achieve a dose response range of 0.03 ng/mL-250 ng/mL.
  • DMSO is added as a negative control. Plates are allowed to pre-incubate for 2 hours before stimulation with 1 ⁇ g/mL anti-CD3 (OKT3; eBioscience). After 72 h, the medium in each well is replaced with 150 ⁇ L of fresh medium containing 10 ⁇ M BrdU (Roche). After 16 h, the supernatant is removed, the plate is dried and the cells fixed by adding 100 ⁇ L of fix/denature solution to each well for 20 min as per the manufacturer's instructions (Roche).
  • Plates are washed once with PBS before addition of the anti-BrdU detection antibody and incubated for 90 mins at room temperature. Plates are then washed gently 3 ⁇ with the wash buffer supplied and developed by addition of 100 ⁇ L of substrate solution. The reaction is stopped by addition of 50 ⁇ L of 1 M H 2 SO 4 , and read for absorbance at 450 nm on a plate reader (Varioskan® Flash, ThermoFisher Scientific). The IC 50 is determined from the dose response curve.
  • Lamina limbal mononuclear cells are isolated and purified from inflamed IBD mucosa of surgical specimens or from normal mucosa of surgical specimens as follows: The mucosa is removed from the deeper layers of the surgical specimens with a scalpel and cut in fragments 3-4 mm size. The epithelium is removed by washing the tissue fragments three times with 1 mM EDTA (Sigma-Aldrich, Poole, UK) in HBSS (Sigma-Aldrich) with agitation using a magnetic stirrer, discarding the supernatant after each wash.
  • 1 mM EDTA Sigma-Aldrich, Poole, UK
  • HBSS Sigma-Aldrich
  • the sample is subsequently treated with type 1A collagenase (1 mg/mL; Sigma-Aldrich) for 1 h with stirring at 37° C.
  • the resulting cell suspension is then filtered using a 100 ⁇ m cell strainer, washed twice, resuspended in RPMI-1640 medium (Sigma-Aldrich) containing 10% fetal calf serum, 100 U/mL penicillin and 100 ⁇ g/mL streptomycin, and used for cell culture.
  • Myofibroblasts from inflamed IBD mucosa are isolated as follows: The mucosa is dissected and discarded and 1 mm-sized mucosal samples are cultured at 37° C. in a humidified CO 2 incubator in Dulbecco's modified Eagle's medium (DMEM, Sigma-Aldrich) supplemented with 20% FBS, 1% non-essential amino acids (Invitrogen, Paisley, UK), 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 50 ⁇ g/mL gentamycin, and 1 ⁇ g/mL amphotericin (Sigma-Aldrich). Established colonies of myofibroblasts are seeded into 25-cm 2 culture flasks and cultured in DMEM supplemented with 20% FBS and antibiotics to at least passage 4 to provide a sufficient quantity for use in stimulation experiments.
  • DMEM Dulbecco's modified Eagle's medium
  • Subconfluent monolayers of myofibroblasts are then seeded in 12-well plates at 3 ⁇ 10 5 cells per well are starved in serum-free medium for 24 h at 37° C., 5% CO 2 before being cultured for 24 h in the presence of either DMSO control or appropriate concentrations of compound. After 24 h the supernatant is removed and assayed for the presence of IL-8 and IL-6 by R&D ELISA. Percentage inhibition of cytokine release by the test compounds is calculated relative to the cytokine release determined for the DMSO control (100%).
  • Neutrophils are isolated from human peripheral blood as follows:
  • Blood is collected by venepuncture and anti-coagulated by addition of 1:1 EDTA: sterile phosphate buffered saline (PBS, no Ca+/Mg+).
  • Dextran 3% w/v is added (1 part dextran solution to 4 parts blood) and the blood allowed to stand for approximately 20 minutes at rt.
  • the supernatant is carefully layered on a density gradient (Lymphoprep, Axis-Shield Healthcare) and centrifuged (15 mins, 2000 rpm, no brake). The supernatant is aspirated off and the cell pellet is re-suspended in sterile saline (0.2%) for no longer than 60 seconds (to lyse contaminating red blood cells).
  • HBSS+ Hops buffered salt solution (without phenol red) containing cytochalasin B (5 ⁇ g/mL) and 1 mM CaCl 2 ) to achieve 5 ⁇ 10 6 cells/mL.
  • test compound 0.3-1000 ng/mL
  • vehicle DMSO, 0.5% final conc
  • Degranulation is stimulated by addition of fMLP (final conc 1 ⁇ M) which after a further incubation (30 mins, 37° C.) the cells are removed by centrifugation (5 mins, 1500 rpm) and the supernatants transferred to a flat bottom 96 well plate.
  • TMB tetramethylbenzidine
  • TK6 cells (lymphoblastic T cell line) are added to the appropriate number of wells of a 96 well plate in 195 ⁇ L of media (RPMI supplemented with 10% foetal bovine serum).
  • 5 ⁇ L of DMSO control (final concentration 0.5% v/v) or test compound (final concentration either 5 or 1 ⁇ g/mL) is added to the wells and incubated at 37° C., 5% CO 2 . After 24 hours, the plate is centrifuged at 1300 rpm for 3 minutes and the supernatant discarded. Cells are then resuspended in 7.5 ⁇ g/mL propidium iodide (PI) in PBS. After 15 minutes, cells are analysed by flow cytometry (BD accuri). The % viability is calculated as the % of cells that are PI negative in the test wells normalised to the DMSO control.
  • PI propidium iodide
  • Non-fasted Balb/c mice are dosed by the intra tracheal route with either vehicle, or the test substance at the indicated times (within the range 2-8 hr) before stimulation of the inflammatory response by application of an LPS challenge.
  • mice are placed into an exposure chamber and exposed to LPS (7.0 mL, 0.5 mg/mL solution in PBS) for 30 min.
  • LPS 7.0 mL, 0.5 mg/mL solution in PBS
  • the animals are anesthetized, their tracheas cannulated and BALF extracted by infusing and then withdrawing from their lungs 1.0 mL of PBS via the tracheal catheter.
  • Total and differential white cell counts in the BALF samples are measured using a Neubaur haemocytometer.
  • Cytospin smears of the BALF samples are prepared by centrifugation at 200 rpm for 5 min at RT and stained using a DiffQuik stain system (Dade Behring). Cells are counted using oil immersion microscopy. Data for neutrophil numbers in BAL are shown as mean ⁇ S.E.M. (standard error of the mean). The percentage inhibition of neutrophil accumulation is calculated for each treatment relative to vehicle treatment.
  • mice Males, 5 weeks old are exposed to cigarette smoke (4% cigarette smoke, diluted with air) for 30 min/day for 11 days using a Tobacco Smoke Inhalation Experiment System for small animals (Model SIS-CS; Sibata Scientific Technology, Tokyo, Japan). Test substances are administered intra-nasally (35 ⁇ L of solution in 50% DMSO/PBS) once daily for 3 days after the final cigarette smoke exposure. At 12 hr after the last dosing, each of the animals is anesthetized, the trachea cannulated and bronchoalveolar lavage fluid (BALF) is collected.
  • BALF bronchoalveolar lavage fluid
  • alveolar macrophages and neutrophils are determined by FACS analysis (EPICS® ALTRA II, Beckman Coulter, Inc., Fullerton, Calif., USA) using anti-mouse MOMA2 antibody (macrophage) or anti-mouse 7/4 antibody (neutrophil).
  • Non-fasted, 10-12 week old, male BDF1 mice are dosed by oral gavage twice daily with either vehicle, reference item (5-ASA) or test compound one day before (Day ⁇ 1) stimulation of the inflammatory response by treatment with dextran sodium sulphate (DSS).
  • DSS dextran sodium sulphate
  • BID dosing of the vehicle 5 mL/kg
  • reference 100 mg/kg
  • test compound 5 mg/kg
  • the drinking water with DSS is replenished every 3 days.
  • During the study animals are weighed every day and stool observations are made and recorded as a score, based on stool consistency.
  • the large intestine is removed and the length and weight are recorded. Sections of the colon are taken for either MPO analysis to determine neutrophil infiltration or for histopathology scoring to determine disease severity.
  • Non-fasted, 10-12 week old, male BDF1 mice are dosed by oral gavage twice daily with either vehicle (5 mL/kg), reference item (Budesonide 2.5 mg/kg) or test compound (1 or 5 mg/kg) one day before (Day ⁇ 1) stimulation of the inflammatory response by treatment with 2,4,6-trinitrobenzenesulphonic acid (TNBS) (15 mg/mL in 50% ethanol/50% saline).
  • TNBS 2,4,6-trinitrobenzenesulphonic acid
  • TNBS 2,4,6-trinitrobenzenesulphonic acid
  • mice are terminated and spleens obtained for CD45RB high cell isolation (Using SCID IBD cell Separation protocol). Approximately 4 ⁇ 10 5 cells/mL CD45RB high cells are then injected IP (100 ⁇ L/mouse) into female SCID animals. On study day 14, mice are weighed and randomized into treatment groups based on body weight. On Day 21 compounds are administered BID, via oral gavage, in a peanut oil vehicle at the dose levels outlined below and a dose volume of 5 mL/kg. Treatment continues until study day 42, at which point the animals are necropsied 4 hours after am administration. The colon length and weight is recorded and used as a secondary endpoint in the study as a measurement of colon oedema.
  • the colon is then divided into six cross-sections, four of which are used for histopathology scoring (primary endpoint) and two are homogenised for cytokine analysis. Data shown is the % inhibition of the induction window between na ⁇ ve animals and vehicle animals, where higher inhibition implies closer to the non-diseased, na ⁇ ve, phenotype.
  • the IC 50 for inhibition of TNF ⁇ release was also measured in PBMCs (using assay (b) above) for the compound of Example 1, and found to be 2.5 nM.
  • the compound of Examples 2 and 13 were also screened in in vivo assay (iv) above, as conducted over 2 days. Histopathology analysis revealed that the compound of Examples 2 and 13 displayed significant activity in this in vivo model of colonic inflammation. In particular, those compounds, when dosed orally both at 5 mg/kg (or, for the compound of Example 2, at 1 and at 5 mg/kg), demonstrated marked improvements in ulcer grade and epithelial repair compared to the vehicle control. In addition, the compound of Examples 2 and 13 produced a marked reduction in inflammatory cell infiltrate in the reticular and lamina limba zones.
  • Examples 2 and 13 were also screened in cellular assay (I), i.e., the ex-vivo human biopsy model described above, where it demonstrated significant anti-inflammatory effects in biopsies from ulcerative colitis (UC) patients.
  • I cellular assay
  • intestinal mucosal biopsies from UC patients have been shown to spontaneously release pro-inflammatory cytokines in vitro (Onken, J. E. et al., J Clin Immunol, 2008, 126(3): 345-352).
  • the compound of Examples 2 and 13 significantly inhibited cytokine (IL-1 ⁇ , IL-6 and IL-8) release compared to the DMSO control when incubated, at 1 ⁇ g/mL, for 24 hours with biopsies from ulcerative colitis patients.
  • cytokine IL-1 ⁇ , IL-6 and IL-8
  • n-, s-, i-, t- and tert- have their usual meanings: normal, secondary, iso, and tertiary.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US14/424,627 2012-08-29 2013-08-28 Kinase inhibitors Abandoned US20150225373A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB1215369.8A GB201215369D0 (en) 2012-08-29 2012-08-29 New compounds
GB1215369.8 2012-08-29
GB1304777.4 2013-03-15
GB201304777A GB201304777D0 (en) 2013-03-15 2013-03-15 New compounds
PCT/GB2013/052251 WO2014033447A2 (fr) 2012-08-29 2013-08-28 Inhibiteurs de kinase

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2013/052251 A-371-Of-International WO2014033447A2 (fr) 2012-08-29 2013-08-28 Inhibiteurs de kinase

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/225,377 Continuation US9790209B2 (en) 2012-08-29 2016-08-01 Kinase inhibitors

Publications (1)

Publication Number Publication Date
US20150225373A1 true US20150225373A1 (en) 2015-08-13

Family

ID=49118571

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/424,627 Abandoned US20150225373A1 (en) 2012-08-29 2013-08-28 Kinase inhibitors
US15/225,377 Active US9790209B2 (en) 2012-08-29 2016-08-01 Kinase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/225,377 Active US9790209B2 (en) 2012-08-29 2016-08-01 Kinase inhibitors

Country Status (3)

Country Link
US (2) US20150225373A1 (fr)
EP (1) EP2890695A2 (fr)
WO (1) WO2014033447A2 (fr)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160039797A1 (en) * 2013-04-02 2016-02-11 Topivert Pharma Limited Kinase inhibitors based upon n-alkyl pyrazoles
US9447076B2 (en) 2014-02-14 2016-09-20 Respivert Ltd. Inhibitor of p38 map kinase
US9481648B2 (en) 2013-04-02 2016-11-01 Respivert Limited Kinase inhibitors
US9499486B2 (en) 2014-10-01 2016-11-22 Respivert Limited Kinase inhibitor
US9701670B2 (en) 2012-08-17 2017-07-11 Respivert Limited Pyrazolyl-ureas as kinase inhibitors
US9732063B2 (en) 2012-11-16 2017-08-15 Respivert Limited Kinase inhibitors
US9783556B2 (en) 2012-08-29 2017-10-10 Respivert Limited Kinase inhibitors
US9796742B2 (en) 2012-08-29 2017-10-24 Respivert Limited Kinase inhibitors
US9850233B2 (en) 2013-03-14 2017-12-26 Respivert Limited Kinase inhibitors
US9890185B2 (en) 2013-12-20 2018-02-13 Respivert Limited Urea derivatives useful as kinase inhibitors
US10238658B2 (en) 2011-10-03 2019-03-26 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-1-yl) ureas as p38 MAP kinase inhibitors
US10266519B2 (en) 2011-10-03 2019-04-23 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-I-yl) ureas as P38 mapkinase inhibitors
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
CN113677674A (zh) * 2019-04-11 2021-11-19 詹森药业有限公司 含有吡啶环的作为malt1抑制剂的衍生物
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5511680B2 (ja) 2007-12-19 2014-06-04 キャンサー・リサーチ・テクノロジー・リミテッド ピリド[2,3−b]ピラジン−8−置換化合物及びその使用
CN104945401B (zh) 2010-02-01 2017-09-05 癌症研究技术有限公司 Ip化合物及它们在治疗中的应用
GB201215357D0 (en) 2012-08-29 2012-10-10 Respivert Ltd Compounds
GB201320729D0 (en) 2013-11-25 2014-01-08 Cancer Rec Tech Ltd Therapeutic compounds and their use
GB201320732D0 (en) 2013-11-25 2014-01-08 Cancer Rec Tech Ltd Methods of chemical synthesis
EA201892213A1 (ru) 2016-04-06 2019-05-31 Топайверт Фарма Лимитед Ингибиторы киназ

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003072569A1 (fr) * 2002-02-25 2003-09-04 Boehringer Ingelheim Pharmaceuticals, Inc. Composes de cycloalkyle uree benzofusionnes a disubstitution en position 1,4 utiles dans le traitement de maladies mediees par la cytokine
WO2006072589A2 (fr) * 2005-01-10 2006-07-13 Novartis Ag Urees disubstituees utilisees en tant qu'inhibiteurs de kinase

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2308428A1 (fr) 1997-11-03 1999-05-14 Boehringer Ingelheim Pharmaceuticals, Inc. Composes heterocycliques aromatiques convenant comme anti-inflammatoires
EP1041982B1 (fr) * 1997-12-22 2011-10-19 Bayer HealthCare LLC Inhibition de l'activite de p38 kinase au moyen d'urees heterocycliques substituees
ATE556713T1 (de) 1999-01-13 2012-05-15 Bayer Healthcare Llc Omega-carboxyarylsubstituierte-diphenyl- harnstoffe als p38-kinasehemmer
EP1140840B1 (fr) 1999-01-13 2006-03-22 Bayer Pharmaceuticals Corp. Diphenylurees a substituants -g(v)-carboxyaryles, inhibitrices de kinase raf
UA73492C2 (en) 1999-01-19 2005-08-15 Aromatic heterocyclic compounds as antiinflammatory agents
JP4820488B2 (ja) 1999-03-12 2011-11-24 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド 抗炎症薬として有用な化合物
ATE312823T1 (de) 1999-07-09 2005-12-15 Boehringer Ingelheim Pharma Verfahren zur herstellung heteroarylsubstituierter ureaverbindungen
MXPA02004594A (es) 1999-11-16 2002-10-23 Boehringer Ingelheim Pharma Derivados de urea como agentes anti-inflamatorios.
US6525046B1 (en) 2000-01-18 2003-02-25 Boehringer Ingelheim Pharmaceuticals, Inc. Aromatic heterocyclic compounds as antiinflammatory agents
US6492529B1 (en) 2000-01-18 2002-12-10 Boehringer Ingelheim Pharmaceuticals, Inc. Bis pyrazole-1H-pyrazole intermediates and their synthesis
WO2002083628A1 (fr) 2001-04-13 2002-10-24 Boehringer Ingelheim Pharmaceuticals, Inc. Composes benzo-fusionnes a disubstitution en positions 1,4
CA2445003A1 (fr) 2001-05-16 2002-11-21 Boehringer Ingelheim Pharmaceuticals, Inc. Derives de diaryluree utilisables en tant qu'agents anti-inflammatoires
ES2284887T3 (es) 2001-07-11 2007-11-16 Boehringer Ingelheim Pharmaceuticals Inc. Metodos para tratar enfermedades transmitidas por citocinas.
AU2003210969A1 (en) 2002-02-11 2003-09-04 Bayer Corporation Aryl ureas with raf kinase and angiogenesis inhibiting activity
AU2003209116A1 (en) 2002-02-11 2003-09-04 Bayer Pharmaceuticals Corporation Aryl ureas with angiogenesis inhibiting activity
WO2004113352A1 (fr) 2003-06-19 2004-12-29 Amedis Pharmaceuticals Ltd. Derives d'heterocyclyluree silyles inhibiteurs des cytokines
CN102060806A (zh) 2003-09-11 2011-05-18 iTherX药品公司 细胞因子抑制剂
WO2005044825A1 (fr) 2003-10-21 2005-05-19 Amedis Pharmaceuticals Ltd. Composes de silicium et utilisations de ceux-ci
GB0423554D0 (en) 2004-10-22 2004-11-24 Cancer Rec Tech Ltd Therapeutic compounds
TWI325423B (en) 2005-10-28 2010-06-01 Lilly Co Eli Kinase inhibitors
GB0818033D0 (en) 2008-10-02 2008-11-05 Respivert Ltd Novel compound
WO2010038086A2 (fr) 2008-10-02 2010-04-08 Respivert Limited Nouveaux composés
AU2009326148B2 (en) 2008-12-11 2014-05-08 Respivert Limited p38 MAP kinase inhibitors
GB0905955D0 (en) 2009-04-06 2009-05-20 Respivert Ltd Novel compounds
GB0921731D0 (en) 2009-12-11 2010-01-27 Respivert Ltd Theraputic uses
GB0921730D0 (en) 2009-12-11 2010-01-27 Respivert Ltd Method of treatment
US8816069B2 (en) 2010-03-15 2014-08-26 Ube Industries, Ltd. Method for producing amide compound
GB201005589D0 (en) 2010-04-01 2010-05-19 Respivert Ltd Novel compounds
EP2556068B1 (fr) 2010-04-08 2019-01-23 Respivert Limited Inhibiteurs de la map kinase p38
US9260410B2 (en) 2010-04-08 2016-02-16 Respivert Ltd. P38 MAP kinase inhibitors
GB201009731D0 (en) 2010-06-10 2010-07-21 Pulmagen Therapeutics Inflamma Kinase inhibitors
GB201010196D0 (en) 2010-06-17 2010-07-21 Respivert Ltd Methods
US8933228B2 (en) 2010-06-17 2015-01-13 Respivert, Ltd. Respiratory formulations and compounds for use therein
GB201010193D0 (en) 2010-06-17 2010-07-21 Respivert Ltd Medicinal use
AU2012320300C1 (en) 2011-10-03 2017-06-01 Respivert Limited 1-Pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl)oxy)napththalen-1-yl) ureas as P38 MAP kinase inhibitors
EP2578582A1 (fr) 2011-10-03 2013-04-10 Respivert Limited 1-Pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl)oxy)napthtalèn-1-yl) urées comme inhibiteurs de la p38 MAP kinase
KR101994381B1 (ko) 2011-12-09 2019-06-28 키에시 파르마슈티시 엣스. 피. 에이. 키나아제 억제제
GB201214750D0 (en) 2012-08-17 2012-10-03 Respivert Ltd Compounds
WO2014033446A1 (fr) 2012-08-29 2014-03-06 Respivert Limited Inhibiteurs de kinases
GB201215357D0 (en) 2012-08-29 2012-10-10 Respivert Ltd Compounds
US9783556B2 (en) 2012-08-29 2017-10-10 Respivert Limited Kinase inhibitors
EP2925742B1 (fr) 2012-11-16 2016-10-26 Respivert Limited Inhibiteurs de kinase
US20160016934A1 (en) 2013-03-14 2016-01-21 Respivert Limited Kinase inhibitors
US9771353B2 (en) 2013-04-02 2017-09-26 Topivert Pharma Limited Kinase inhibitors based upon N-alkyl pyrazoles
EP2981526B1 (fr) 2013-04-02 2021-11-03 Oxular Acquisitions Limited Produits intermédiares utiles pour la synthèse de dérivés naphthyl-urée en tant qu'inhibiteurs de kinase
MX371353B (es) 2014-02-14 2020-01-27 Respivert Ltd Compuestos heterociclicos aromaticos como compuestos antiinflamatorios.
MA40774A (fr) 2014-10-01 2017-08-08 Respivert Ltd Dérivés de diaryle-urée en tant qu'inhibiteurs de kinase p38

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003072569A1 (fr) * 2002-02-25 2003-09-04 Boehringer Ingelheim Pharmaceuticals, Inc. Composes de cycloalkyle uree benzofusionnes a disubstitution en position 1,4 utiles dans le traitement de maladies mediees par la cytokine
WO2006072589A2 (fr) * 2005-01-10 2006-07-13 Novartis Ag Urees disubstituees utilisees en tant qu'inhibiteurs de kinase

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
A.C. Brando et al., 63 Pharmacological Reports, 1029-1039 (2011) *
C.L. Sawyers, Nature, 548-552 (2008) *
C.M. Coughlin et al., Breast Cancer Research Treatment, 1-11 (2010) *
D. Singh et al., 50 The Journal of Clinical Pharmacology, 94-100 (2010) *
E.S. Masuda et al., 21 Pulmonary Pharmacology & Therapeutics, 461-467 (2008) *
G. Liu et al., 31 Arteriosclerosis, Thrombosis and Vascular Biology, 1342-1350 (2011) *
KINASE INHIBITORS, METHODS IN MOLECULAR BIOLOGY 795 (B. Kuster ed., 2012) *
M.E. Weinblatt et al., 363 The New England Journal of Medicine 1303-1312 (2010) *
M.P. Kim et al., 335 Cell and Tissue Research, 249-259 (2009) *
N. Yamamoto et al., 306 The Journal Of Pharmacology And Experimental Therapeutics, 1174-1181 (2003) *
R. Singh et al., 42 Annual Reports in Medicinal Chemistry, 379-391 (2007) *
R.S. Jope et al., 32 Neurochemical Research, 577-595 (2007) *
U. McDermott et al., 27 Journal of Clinical Oncology, 5650-5659 (2009) *

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10813932B2 (en) 2011-10-03 2020-10-27 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-1-yl) ureas as P38 MAP knase inhibitors
US10738032B2 (en) 2011-10-03 2020-08-11 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-i-yl) ureas as P38 mapkinase inhibitors
US10266519B2 (en) 2011-10-03 2019-04-23 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-I-yl) ureas as P38 mapkinase inhibitors
US10238658B2 (en) 2011-10-03 2019-03-26 Respivert Limited 1-pyrazolyl-3-(4-((2-anilinopyrimidin-4-yl) oxy) napththalen-1-yl) ureas as p38 MAP kinase inhibitors
US9701670B2 (en) 2012-08-17 2017-07-11 Respivert Limited Pyrazolyl-ureas as kinase inhibitors
US9796742B2 (en) 2012-08-29 2017-10-24 Respivert Limited Kinase inhibitors
US9783556B2 (en) 2012-08-29 2017-10-10 Respivert Limited Kinase inhibitors
US9732063B2 (en) 2012-11-16 2017-08-15 Respivert Limited Kinase inhibitors
US10301288B2 (en) 2013-03-14 2019-05-28 Topivert Pharma Limited Kinase inhibitors
US9850233B2 (en) 2013-03-14 2017-12-26 Respivert Limited Kinase inhibitors
US10435361B2 (en) 2013-04-02 2019-10-08 Topivert Pharma Limited Kinase inhibitors
US20160039797A1 (en) * 2013-04-02 2016-02-11 Topivert Pharma Limited Kinase inhibitors based upon n-alkyl pyrazoles
US9790174B2 (en) 2013-04-02 2017-10-17 Respivert Limited Kinase inhibitors
US9771353B2 (en) * 2013-04-02 2017-09-26 Topivert Pharma Limited Kinase inhibitors based upon N-alkyl pyrazoles
US9481648B2 (en) 2013-04-02 2016-11-01 Respivert Limited Kinase inhibitors
US9890185B2 (en) 2013-12-20 2018-02-13 Respivert Limited Urea derivatives useful as kinase inhibitors
US11142515B2 (en) 2014-02-14 2021-10-12 Respivert Limited Pyrazolyl-ureas as kinase inhibitors
US10045980B2 (en) 2014-02-14 2018-08-14 Respivert Ltd Inhibitor of p38 map kinase
US10815217B2 (en) 2014-02-14 2020-10-27 Respivert Ltd. Inhibitor of p38 MAP kinase
US9624196B2 (en) 2014-02-14 2017-04-18 Respivert Ltd. Inhibitor of p38 MAP kinase
US10294216B2 (en) 2014-02-14 2019-05-21 Respivert Limited Pyrazolyl ureas as kinase inhibitors
US9447076B2 (en) 2014-02-14 2016-09-20 Respivert Ltd. Inhibitor of p38 map kinase
US9884845B2 (en) 2014-02-14 2018-02-06 Respivert Limited Pyrazolyl-ureas as kinase inhibitors
US11634406B2 (en) 2014-02-14 2023-04-25 Respivert Ltd. Inhibitor of p38 MAP kinase
US10392346B2 (en) 2014-10-01 2019-08-27 Topivert Pharma Limited Kinase inhibitors
US9499486B2 (en) 2014-10-01 2016-11-22 Respivert Limited Kinase inhibitor
US9751837B2 (en) 2014-10-01 2017-09-05 Respivert Limited Kinase inhibitors
US10941115B2 (en) 2014-10-01 2021-03-09 Oxular Acquisitions Limited Kinase inhibitors
US10125100B2 (en) 2014-10-01 2018-11-13 Respivert Limited Kinase inhibitors
US9822076B2 (en) 2014-10-01 2017-11-21 Respivert Limited Kinase inhibitor
US10537560B2 (en) 2017-10-05 2020-01-21 Fulcrum Therapeutics. Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11479770B2 (en) 2017-10-05 2022-10-25 Fulcrum Therapeutics, Inc. Use of p38 inhibitors to reduce expression of DUX4
CN113677674A (zh) * 2019-04-11 2021-11-19 詹森药业有限公司 含有吡啶环的作为malt1抑制剂的衍生物

Also Published As

Publication number Publication date
EP2890695A2 (fr) 2015-07-08
US20160340343A1 (en) 2016-11-24
WO2014033447A3 (fr) 2014-09-18
US9790209B2 (en) 2017-10-17
WO2014033447A2 (fr) 2014-03-06

Similar Documents

Publication Publication Date Title
US9790209B2 (en) Kinase inhibitors
US9783556B2 (en) Kinase inhibitors
US9796742B2 (en) Kinase inhibitors
US10301288B2 (en) Kinase inhibitors
US10435361B2 (en) Kinase inhibitors
US9771353B2 (en) Kinase inhibitors based upon N-alkyl pyrazoles
US9701670B2 (en) Pyrazolyl-ureas as kinase inhibitors
US9732063B2 (en) Kinase inhibitors
US9890185B2 (en) Urea derivatives useful as kinase inhibitors
US20170209445A1 (en) Kinase inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: TOPIVERT PHARMA LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FYFE, MATTHEW COLIN THOR;MEGHANI, PREMJI;STENT, MATTHEW ALEXANDER HENRY;AND OTHERS;SIGNING DATES FROM 20150206 TO 20150225;REEL/FRAME:035117/0389

Owner name: RESPIVERT LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FYFE, MATTHEW COLIN THOR;MEGHANI, PREMJI;STENT, MATTHEW ALEXANDER HENRY;AND OTHERS;SIGNING DATES FROM 20150206 TO 20150225;REEL/FRAME:035117/0389

STCB Information on status: application discontinuation

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