WO2010010188A1 - Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires - Google Patents

Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires Download PDF

Info

Publication number
WO2010010188A1
WO2010010188A1 PCT/EP2009/059600 EP2009059600W WO2010010188A1 WO 2010010188 A1 WO2010010188 A1 WO 2010010188A1 EP 2009059600 W EP2009059600 W EP 2009059600W WO 2010010188 A1 WO2010010188 A1 WO 2010010188A1
Authority
WO
WIPO (PCT)
Prior art keywords
unsubstituted
substituted
compound
alkyl
pharmaceutically acceptable
Prior art date
Application number
PCT/EP2009/059600
Other languages
English (en)
Inventor
Christel Jeanne Marie Menet
Javier Blanc
Nolwenn Jouannigot
Alastair James Hodges
Luc Juliaan Corina Van Rompaey
Stephen Robert Fletcher
Original Assignee
Galapagos Nv
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Galapagos Nv filed Critical Galapagos Nv
Publication of WO2010010188A1 publication Critical patent/WO2010010188A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis

Definitions

  • the present invention relates to compounds that are inhibitors of JAK, a family of tyrosine kinases that are involved in the modulation of the degradation of cartilage, joint degeneration and diseases involving such degradation and/or inflammation.
  • the present invention also provides methods for the production of these compounds, pharmaceutical compositions comprising these compounds, methods for the prevention and/or treatment of diseases involving cartilage degradation, bone and/or joint degradation, conditions involving inflammation or immune responses, endotoxin- driven disease states, cancer, and organ transplant rejection; and/ or methods for the prevention and/or treatment of diseases involving cartilage degradation, joint degradation and/or inflammation by administering a compound of the invention.
  • Janus kinases are cytoplasmic tyrosine kinases that transduce cytokine signaling from membrane receptors to STAT transcription factors.
  • JAK family members Four JAK family members are described, JAKl, JAK2, JAK3 and TYK2.
  • JAK family members Upon binding of the cytokine to its receptor, JAK family members auto- and/or transphosphorylate each other, followed by phosphorylation of STATs that then migrate to the nucleus to modulate transcription.
  • JAK-STAT intracellular signal transduction serves the interferons, most interleukins, as well as a variety of cytokines and endocrine factors such as EPO, TPO, GH, OSM, LIF, CNTF, GM-CSF, PRL Vainchenker W. et al (2008).
  • JAK3 is validated by mouse and human genetics as an immune- suppression target (O'Shea J. et al. (2004)). JAK3 inhibitors were successfully taken into clinical development, initially for organ transplant rejection but later also in other immuno-inflammatory indications such as rheumathoid arthritis (RA), psoriasis and Crohn's disease (http://clinicaltrials.gov/).
  • RA rheumathoid arthritis
  • psoriasis http://clinicaltrials.gov/.
  • TYK2 is a potential target for immuno-inflammatory diseases, being validated by human genetics and mouse knock-out studies (Levy D. and Loomis C. (2007)).
  • JAKl is a novel target in the immuno-inflammatory disease area. JAKl heterodimerizes with the other JAKs to transduce cytokine- driven pro-inflammatory signaling. Therefore, inhibition of JAKl and/or other JAKs is expected to be of therapeutic benefit for a range of inflammatory conditions as well as for other diseases driven by JAK-mediated signal transduction.
  • Cartilage is an avascular tissue of which chondrocytes are the main cellular component.
  • the chondrocytes in normal articular cartilage occupy approximately 5% of the tissue volume, while the extra- cellular matrix makes up the remaining 95% of the tissue.
  • the chondrocytes secrete the components of the matrix, mainly proteoglycans and collagens, which in turn supply the chondrocytes with an environment suitable for their survival under mechanical stress.
  • collagen type II together with the protein collagen type IX, is arranged in solid fibril-like structures which provide cartilage with great mechanical strength.
  • the proteoglycans can absorb water and are responsible for the resilient and shock absorbing properties of the cartilage.
  • cartilage degradation is caused by the secretion of proteases (e.g. collagenases) by inflamed tissues (the inflamed synovium for example).
  • cartilage degradation can also be the result of an injury of the cartilage, due to an accident or surgery, or exaggerated loading or 'wear and tear'.
  • the ability of cartilage tissue to regenerate after such insults is limited. Chondrocytes in injured cartilage often display reduced cartilage synthesizing (anabolic) activity and / or increased cartilage degrading (catabolic) activity.
  • RA Rheumatoid arthritis
  • osteoarthritis are the most prominent.
  • RA Rheumatoid arthritis
  • the aim of an RA therapy is not only to slow down the disease but to attain remission in order to stop the joint destruction.
  • the high prevalence of RA ⁇ 0.8% of the adults are affected worldwide
  • Osteoarthritis also referred to as OA, or wear-and-tear arthritis
  • OA wear-and-tear arthritis
  • the disease mainly affects hands and weight-bearing joints such as knees, hips and spines. This process thins the cartilage. When the surface area has disappeared due to the thinning, a grade I osteoarthritis is reached; when the tangential surface area has disappeared, grade II osteoarthritis is reached.
  • Osteoarthritis is difficult to treat. At present, no cure is available and treatment focuses on relieving pain and preventing the affected joint from becoming deformed. Common treatments include the use of non-steroidal anti-inflammatory drugs (NSAIDs). Although dietary supplements such as chondroitin and glucosamine sulphate have been advocated as safe and effective options for the treatment of osteoarthritis, a recent clinical trial revealed that both treatments did not reduce pain associated to osteoarthritis. (Clegg et ah,
  • chondral cellular material is taken from the patient, sent to a laboratory where it is expanded. The material is then implanted in the damaged tissues to cover the tissue's defects.
  • Another treatment includes the intra-articular instillation of Hylan G-F 20 (e.g. Synvisc®,
  • Hyalgan®, Artz® a substance that improves temporarily the rheology of the synovial fluid, producing an almost immediate sensation of free movement and a marked reduction of pain.
  • Stimulation of the anabolic processes, blocking catabolic processes, or a combination of these two, may result in stabilization of the cartilage, and perhaps even reversion of the damage, and therefore prevent further progression of the disease.
  • Various triggers may stimulate anabolic stimulation of chondrocytes.
  • IGF-I Insulin-like growth factor-I
  • BMP bone morphogenetic protein
  • TGF- ⁇ human transforming growth factor- ⁇
  • JAKl belongs to the Janus kinase (JAK) family of cytoplasmic tyrosine kinases, involved in cytokine receptor-mediated intracellular signal transduction.
  • the JAK family consists of 4 members: JAKl, JAK2, JAK3 and TYK2. JAKs are recruited to cytokine receptors, upon binding of the cytokine, followed by heterodimerization of the cytokine receptor and a shared receptor subunit (common gamma-c chain, gpl30).
  • JAKs are then activated by auto- and/or transphosphorylation by another JAK, resulting in phosphorylation of the receptors and recruitment and phosphorylation of members of the signal transducer and activator of transcription (STATs).
  • STATs signal transducer and activator of transcription
  • Phosphorylated STATs dimerize and translocate to the nucleus where they bind to enhancer regions of cytokine-responsive genes.
  • Knockout of the JAKl gene in mice demonstrated that JAKl plays essential and nonredundant roles during development: JAKl-/- mice died within 24h after birth and lymphocyte development was severely impaired.
  • JAKl -/- cells were not, or less, reactive to cytokines that use class II cytokine receptors, cytokine receptors that use the gamma-c subunit for signaling and the family of cytokine receptors that use the gpl30 subunit for signaling (Rodig et al., 1998).
  • JAK family members have been implicated in additional conditions including myeloproliferative disorders (O'Sullivan et al, 2007, MoI Immunol. 44(10):2497-506), where mutations in JAK2 have been identified. This indicates that inhibitors of JAK in particular JAK2 may also be of use in the treatment of myeloproliferative disorders. Additionally, the JAK family, in particular JAKl, JAK2 and JAK3, has been linked to cancers, in particular leukaemias e.g. acute myeloid leukaemia (O'Sullivan et al, 2007, MoI Immunol.
  • JAK3 and Tyk2 A link with autoimmune diseases has been established for JAK3 and Tyk2. Mutations in JAK3 but also in the upstream signaling components gamma-c receptor chain and IL7 receptor account in aggregate for -70% of cases of human severe combined immunodeficiency ('OShea et al., 2004). Note that JAKl cooperates with JAK3 in transducing signals from the gamma-c receptor chain. Tyk2 polymorphisms are seen in systemic lupus erythematosus (SLE) (O'Sullivan et al, 2007, MoI Immunol. 44(10):2497-506). Hence, targeting the JAK family may provide a therapeutic opportunity in the immuno-inflammation area.
  • SLE systemic lupus erythematosus
  • the current therapies are not satisfactory and therefore there remains a need to identify further compounds that may be of use in the treatment of diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g.
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers , especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • the present invention therefore provides compounds, methods for their manufacture and a pharmaceutical comprising a compound of the invention together with a suitable pharmaceutical carrier.
  • the present invention also provides for the use of a compound of the invention in the preparation of a medicament for the treatment of degenerative joint diseases.
  • the present invention is based on the discovery that inhibitors of JAK are useful for the treatment of diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g.
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • the present invention also provides methods for the production of these compounds, pharmaceutical compositions comprising these compounds and methods for treating diseases involving cartilage degradation, joint degradation and/or inflammation by administering a compound of the invention.
  • substituted bicycloheteroaryl compounds having according to Formula (I):
  • CyI and Cy2 is independently selected from aryl and heteroaryl
  • Ll is selected from a single bond, -O-, -C(O)-, -S(O) 2 -, -N(R 4a )-, -CON(R 4a )-, -SO 2 N(R 4a )-, - N(R 4a )CO-, or - N(R 4a )SO 2 -; each R 1 is independently selected from Ci-Ce alkyl, substituted Ci-C ⁇ alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted amido, substituted or unsubstituted Ci-Ce alkoxy, substituted or unsubstituted amino, substituted sulfmyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, substituted or unsubstituted C
  • substituted bicycloheteroaryl compounds which are able to inhibit the activity of JAK in vivo according to Formula (I).
  • the present invention provides pharmaceutical compositions comprising a compound of the invention, and a pharmaceutical carrier, excipient or diluent.
  • the pharmaceutical composition can comprise one or more of the compounds described herein.
  • the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein are all pharmaceutically acceptable as prepared and used.
  • this invention provides a method of treating a mammal susceptible to or afflicted with a condition from among those listed herein, and particularly, such condition as may be associated with aberrant JAK activity, for example diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g.
  • a condition from among those listed herein, and particularly, such condition as may be associated with aberrant JAK activity, for example diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis,
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases, which method comprises administering a therapeutically effective amount of a compound of the invention or a pharmceutical compostion as herein described.
  • the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g.
  • the present invention provides a method for treating conditions selected from inflammation, such as rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), inflammatory bowel diseases (e.g. Crohn's disease, colitis), endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), and organ transplant rejection; and cartilage, bone and/or joint degradation or degeneration, such as osteoarthritis, which method comprises administering an effective amount of one or more of the pharmaceutical compositions or compounds herein described.
  • inflammation such as rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), inflammatory bowel diseases (e.g. Crohn's disease, colitis), endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endot
  • the present invention provides a method of treating a mammal susceptible to or afflicted with proliferative disorders in particular cancer, (e.g. solid tumours), leukaemias, multiple myeloma or psoriasis.
  • cancer e.g. solid tumours
  • leukaemias e.g. multiple myeloma or psoriasis.
  • the present invention provides a compound of the invention for use in the treatment or prevention of a condition selected from those listed herein, particularly such conditions as may be associated with aberrant JAK activity such as diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g.
  • a condition selected from those listed herein particularly such conditions as may be associated with aberrant JAK activity such as diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic air
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • the condition is selected from inflammation, such as rheumatoid arthritis, juvenile idiopathic arthritis, psoriasis, allergic airways disease (e.g.
  • asthma wheezing a bowel disease
  • inflammatory bowel diseases e.g. Crohn's disease, colitis
  • endotoxin- driven disease states e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure
  • organ transplant rejection e.g. cartilage, bone and/or joint degradation or degeneration, such as osteoarthritis.
  • the present invention provides a compound of the invention for use in the treatment or prevention of proliferative disorders, in particular cancer, (e.g. solid tumours), leukaemias, multiple myeloma or psoriasis.
  • cancer e.g. solid tumours
  • leukaemias e.g. multiple myeloma or psoriasis.
  • this invention provides a method for treating a mammal susceptible to or afflicted with a condition that is causally related to abnormal JAK activity as described herein, and comprises administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds herein described.
  • the present invention provides a compound of the invention for use in the treatment or prevention of a condition that is causally related to abnormal JAK activity.
  • this invention provides methods for synthesizing the compounds of the invention, with representative synthetic protocols and pathways disclosed later on herein.
  • a still further object of this invention is to provide pharmaceutical compositions that may be used in the treatment or prevention of a variety of disease states, including the diseases associated with JAK activity such as diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g.
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers , especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • the condition is selected from inflammation, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g.
  • asthma wheezing fever
  • rhinitis juvenile idiopathic arthritis
  • colitis inflammatory bowel diseases
  • endotoxin- driven disease states e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure
  • organ transplant rejection e.g., hematomas, hematomas, and hematomas.
  • cartilage, bone and/or joint degradation or degeneration such as osteoarthritis or cancers (e.g. solid tumours or leukaemias).
  • 'Acyl' or 'Alkanoyl' refers to a radical -C(O)R , where R is hydrogen, Ci-C 8 alkyl, C 3 -C 10 cycloalkyl, C 3 -C 10 cycloalkylmethyl, 4-10 membered heterocycloalkyl, aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl as defined herein.
  • Representative examples include, but are not limited to, formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl and benzylcarbonyl.
  • Exemplary 'acyl' groups are -C(O)H, -C(O)-Ci-C 8 alkyl, -C(O)-(CH 2 MC 6 -C 10 aryl), -C(O)-(CH 2 ) t (5-10 membered heteroaryl), -C(O)-(CH 2 XC 3 -Ci 0 cycloalkyl), and -C(O)-(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4.
  • 'Substituted Acyl' or 'Substituted Alkanoyl' refers to a radical -C(O)R 21 , wherein R 21 is independently
  • 'Acylamino' refers to a radical -NR 22 C(O)R 23 , where R 22 is hydrogen, CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, arylalkyl, 5-10 memberd heteroaryl or heteroarylalkyl and R is hydrogen, CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, as defined herein.
  • Exemplary 'acylamino' include, but are not limited to, formylamino, acetylamino, cyclohexylcarbonylamino, cyclohexylmethyl- carbonylamino, benzoylamino and benzylcarbonylamino.
  • Exemplary 'acylamino' groups are -NR 21 C(O)-Cp C 8 alkyl, -NR 21' C(O)-(CH 2 ) t (C 6 -Ci 0 aryl), -NR 21 C(O)-(CH 2 ) t (5-10 membered heteroaryl), -NR 21 C(O)- (CH 2 )t(C 3 -Cio cycloalkyl), and -NR 21 C(O)-(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R 21 independently represents H or CpC 8 alkyl.
  • 'Substituted Acylamino' refers to a radical -NR 24 C(O)R 25 , wherein:
  • R 24 is independently
  • R 5 is independently
  • alkoxy' refers to the group -OR 26 where R 26 is CpCg alkyl.
  • Particular alkoxy groups are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, and 1,2- dimethylbutoxy.
  • Particular alkoxy groups are lower alkoxy, i.e. with between 1 and 6 carbon atoms. Further particular alkoxy groups have between 1 and 4 carbon atoms.
  • Substituted alkoxy' refers to an alkoxy group substituted with one or more of those groups recited in the definition of "substituted” herein, and particularly refers to an alkoxy group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, in particular 1 substituent, selected from the group consisting of amino, substituted amino, C 6 -Ci 0 aryl, -O-aryl, carboxyl, cyano, C3-C 1 0 cycloalkyl, 4-10 membered heterocycloalkyl, halogen, 5-10 membered heteroaryl, hydroxyl, nitro, thioalkoxy, thio-O-aryl, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(O) 2 - and aryl-S(O) 2 -.
  • Exemplary 'substituted alkoxy' groups are -O-(CH 2 ) t (C 6 -Ci 0 aryl), -O-(CH 2 ) t (5-10 membered heteroaryl), -O-(CH 2 ) t (C 3 - Cio cycloalkyl), and -O-(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4 and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, unsubstituted C 1 -C 4 alkoxy, unsubstituted C 1 -C 4 haloalkyl, unsubstituted C 1 -C 4 hydroxyalkyl, or unsubstituted C 1 -C 4 haloalkoxy or hydroxy.
  • 'Alkoxycarbonyl' refers to a radical -C(O)-OR 27 where R 27 represents an CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci O cycloalkylalkyl, 4-10 membered heterocycloalkylalkyl, aralkyl, or 5-10 membered heteroarylalkyl as defined herein.
  • alkoxycarbonyl groups are C(O)O-CpCg alkyl, -C(O)O- (CH 2 MC 6 -C 10 aryl), -C(O)O-(CH 2 ) t (5-10 membered heteroaryl), -C(O)O-(CH 2 MC 3 -C 10 cycloalkyl), and - C(O)O-(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 1 to 4.
  • 'Substituted Alkoxycarbonyl' refers to a radical -C(O)-OR 28 where R 28 represents:
  • 'Alkyl' means straight or branched aliphatic hydrocarbon having 1 to 20 carbon atoms.
  • Particular alkyl has 1 to 12 carbon atoms. More particular is lower alkyl which has 1 to 6 carbon atoms. A further particular group has 1 to 4 carbon atoms.
  • Exemplary straight chained groups include methyl, ethyl n- propyl, and n-butyl. Branched means that one or more lower alkyl groups such as methyl, ethyl, propyl or butyl is attached to a linear alkyl chain, exemplary branched chain groups include isopropyl, iso-butyl, t-butyl and isoamyl.
  • Substituted alkyl' refers to an alkyl group as defined above substituted with one or more of those groups recited in the definition of "substituted” herein, and particularly refers to an alkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, in particular 1 substituent, selected from the group consisting of acyl, acylamino, acyloxy (-O-acyl or - OC(O)R 20 ), alkoxy, alkoxycarbonyl, alkoxycarbonylamino (-NR -alkoxycarbonyl or -NH-C(O)-OR 27 ), amino, substituted amino, aminocarbonyl (carbamoyl or amido or -C(O)-NR 2 ), aminocarbonylamino (-NR -C(O)- NR 2 ), aminocarbonyloxy (-O-C(O)-NR 2) , aminosulfon
  • 'substituted alkyl' refers to a CpC 8 alkyl group substituted with halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR SO 2 R , -SO 2 NR R , -C(O)R , - C(O)OR “ , -OC(O)R “ , -NR “' C(O)R “ , -C(O)NR “ R “ , -NR “ R “ , or -(CR “' R “” ) m OR " ; wherein each R " is independently selected from H, CpC 8 alkyl, -(CH 2 )t(C 6 -Cio aryl), -(CH 2 ) t (5-10 membered heteroaryl), - (CH 2 ) t (C3-Cio cycloalkyl), and -(CH 2 ) t (4-10 membered heterocyclo
  • 'Amino' refers to the radical -NH 2 .
  • 'Substituted amino' refers to an amino group substituted with one or more of those groups recited in the definition of 'substituted' herein, and particularly refers to the group -N(R 33 ) 2 where each R 33 is independently selected from:
  • R 33 groups When both R 33 groups are hydrogen, -N(R 33 ) 2 is an amino group.
  • Exemplary 'substituted amino' groups are -NR 33' -CpC 8 alkyl, -NR 33' -(CH 2 ) t (C 6 -Cio aryl), -NR 33' -(CH 2 ) t (5-10 membered heteroaryl), -NR 33' -(CH 2 ) t (C 3 -Cio cycloalkyl), and -NR 33' -(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R 33 independently represents H or CpCg alkyl; and any alkyl groups present, may themselves be substituted by halo, substituted or unsubstituted amino, or hydroxy; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsub
  • Aminosulfonyl or “Sulfonamide” refers to the radical -S(O 2 )NH 2 .
  • Substituted aminosulfonyl or “substituted sulfonamide” refers to a radical such as -
  • each R 48 is independently selected from:
  • Exemplary 'substituted aminosulfonyl' or 'substituted sulfonamide' groups are -S(O 2 )N(R 48 )-
  • 'Aralkyl' or 'arylalkyl' refers to an alkyl group, as defined above, substituted with one or more aryl groups, as defined above. Particular aralkyl or arylalkyl groups are alkyl groups substituted with one aryl group.
  • 'Substituted Aralkyl' or 'substituted arylalkyl' refers to an alkyl group, as defined above, substituted with one or more aryl groups; and at least one of any aryl group present, may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, cyano, unsubstituted C 1 -C 4 alkoxy, unsubstituted CpC 4 haloalkyl, unsubstituted C 1 -C 4 hydroxyalkyl, or unsubstituted C 1 -C 4 haloalkoxy or hydroxy.
  • 'Aryl' refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system.
  • aryl refers to an aromatic ring structure, mono-cyclic or poly-cyclic that includes from 5 to 12 ring members, more usually 6 to
  • aryl group is a monocyclic ring system it preferentially contains 6 carbon atoms.
  • Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene
  • 'Substituted Aryl' refers to an aryl group substituted with one or more of those groups recited in the definition of 'substituted' herein, and particularly refers to an aryl group that may optionally be substituted with 1 or more substituents, for instance from 1 to 5 substituents, particularly 1 to 3 substituents, in particular 1 substituent.
  • 'Substituted Aryl' refers to an aryl group substituted with one or more of groups selected from halo, CpCg alkyl, CpCg haloalkyl, CpCg haloalkoxy, cyano, hydroxy, CpCg alkoxy, and amino.
  • R 49 and R 50 may be hydrogen and at least one of R 49 and R 50 is each independently selected from CpCg alkyl, 4-10 membered heterocycloalkyl, alkanoyl, CpCg alkoxy, hetero-O- aryl, alkylamino, arylamino, heteroarylamino, NR 51 COR 52 , NR 51 SOR 52 NR 51 SO 2 R 52 , COOalkyl, COOaryl, CONR 51 R 52 , CONR 51 OR 52 , NR 51 R 52 , SO 2 NR 51 R 52 , S-alkyl, SOalkyl, SO 2 alkyl, Saryl, SOaryl, SO 2 aryl; or R 49 and R 50 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionally containing one or more heteroatoms selected from the group N, O or S.
  • R 49 and R 50 may be joined to form a cyclic ring (satur
  • R 51 , and R 52 are independently hydrogen, CpCg alkyl, C 1 -C 4 haloalkyl, C3-C 1 0 cycloalkyl, 4-10 membered heterocycloalkyl, C ⁇ -Cio aryl, substituted aryl, 5-10 membered heteroaryl.
  • Arylalkyloxy' refers to an -O-alkylaryl radical where alkylaryl is as defined herein.
  • Arylalkyloxy refers to an -O-alkylaryl radical where alkylaryl is as defined herein; and any aryl groups present, may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, cyano, unsubstituted C 1 -C 4 alkoxy, unsubstituted Cp 4 haloalkyl, unsubstituted C 1 -C 4 hydroxyalkyl, or unsubstituted C 1 -C 4 haloalkoxy or hydroxy.
  • 'Azido' refers to the radical -N 3 .
  • Carbamoyl or amido' refers to the radical -C(O)NH 2 .
  • R 5 is independently
  • Exemplary 'Substituted Amido / Carbamoyl' groups are -C(O) NR 53' -CpC 8 alkyl, -C(0)NR 53' -(CH 2 ) t (C 6 -Cio aryl), -C(O)N 53' -(CH 2 ) t (5-10 membered heteroaryl), -C(O)NR 53' -(CH 2 ) t (C 3 -Ci 0 cycloalkyl), and -C(O)NR 53' - (CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R 53 independently represents H or CpCg alkyl and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, unsubstituted C 1 -C 4 al
  • 'Cycloalkyl' refers to cyclic non-aromatic hydrocarbyl groups having from 3 to 10 carbon atoms.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • 'Substituted cycloalkyl' refers to a cycloalkyl group as defined above substituted with one or more of those groups recited in the definition of 'substituted' herein, and particularly refers to a cycloalkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, in particular 1 substituent.
  • 'Cyano' refers to the radical -CN.
  • 'Halo' or 'halogen' refers to fluoro (F), chloro (Cl), bromo (Br) and iodo (I). Particular halo groups are either fluoro or chloro.
  • Hetero when used to describe a compound or a group present on a compound means that one or more carbon atoms in the compound or group have been replaced by a nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g. heteroalkyl, cycloalkyl, e.g. heterocycloalkyl, aryl, e.g. heteroaryl, cycloalkenyl, e.g. cycloheteroalkenyl, and the like having from 1 to 5, and particularly from 1 to 3 heteroatoms.
  • Heteroaryl' means an aromatic ring structure, mono-cyclic or polycyclic, that includes one or more heteroatoms and 5 to 12 ring members, more usually 5 to 10 ring members.
  • the heteroaryl group can be, for example, a five membered or six membered monocyclic ring or a bicyclic structure formed from fused five and six membered rings or two fused six membered rings or, by way of a further example, two fused five membered rings.
  • Each ring may contain up to four heteroatoms typically selected from nitrogen, sulphur and oxygen.
  • the heteroaryl ring will contain up to 4 heteroatoms, more typically up to 3 heteroatoms, more usually up to 2, for example a single heteroatom.
  • the heteroaryl ring contains at least one ring nitrogen atom.
  • the nitrogen atoms in the heteroaryl rings can be basic, as in the case of an imidazole or pyridine, or essentially non-basic as in the case of an indole or pyrrole nitrogen. In general the number of basic nitrogen atoms present in the heteroaryl group, including any amino group substituents of the ring, will be less than five.
  • Examples of five membered monocyclic heteroaryl groups include but are not limited to pyrrole, furan, thiophene, imidazole, furazan, oxazole, oxadiazole, oxatriazole, isoxazole, thiazole, isothiazole, pyrazole, triazole and tetrazole groups.
  • Examples of six membered monocyclic heteroaryl groups include but are not limited to pyridine, pyrazine, pyridazine, pyrimidine and triazine.
  • bicyclic heteroaryl groups containing a five membered ring fused to another five membered ring include but are not limited to imidazothiazole and imidazoimidazole.
  • bicyclic heteroaryl groups containing a six membered ring fused to a five membered ring include but are not limited to benzfuran, benzthiophene, benzimidazole, benzoxazole, isobenzoxazole, benzisoxazole, benzthiazole, benzisothiazole, isobenzofuran, indole, isoindole, isoindolone, indolizine, indoline, isoindoline, purine (e.g., adenine, guanine), indazole, pyrazolopyrimidine, triazolopyrimidine, benzodioxole and pyrazolopyridine groups.
  • bicyclic heteroaryl groups containing two fused six membered rings include but are not limited to quinoline, isoquinoline, chroman, thiochroman, chromene, isochromene, chroman, isochroman, benzodioxan, quinolizine, benzoxazine, benzodiazine, pyridopyridine, quinoxaline, quinazoline, cinnoline, phthalazine, naphthyridine and pteridine groups.
  • heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine.
  • Examples of representative aryl having hetero atoms containing substitution include the following: wherein each W is selected from C(R 54 ) 2 , NR 54 , O and S; and each Y is selected from carbonyl, NR 54 , O and S; and R is independently hydrogen, Ci-Cg alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -C 10 aryl, and 5-10 membered heteroaryl.
  • each Y is selected from carbonyl, N, NR 55 , O and S; and R 55 is independently hydrogen, Ci-Cg alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, and 5-10 membered heteroaryl.
  • the term 'heterocycloalkyl' refers to a 4-10 membered, stable heterocyclic non-aromatic ring and/or including rings containing one or more heteroatoms independently selected from N, O and S, fused thereto.
  • a fused heterocyclic ring system may include carbocyclic rings and need only include one heterocyclic ring.
  • heterocyclic rings include, but are not limited to, morpholine, piperidine (e.g. 1-piperidinyl, 2-piperidinyl, 3-piperidinyl and 4-piperidinyl), pyrrolidine (e.g. 1 -pyrrolidinyl, 2- pyrrolidinyl and 3 -pyrrolidinyl), pyrrolidone, pyran (2H-pyran or 4H-pyran), dihydrothiophene, dihydropyran, dihydrofuran, dihydrothiazole, tetrahydrofuran, tetrahydrothiophene, dioxane, tetrahydropyran (e.g.
  • heterocycloalkyl groups are shown in the following illustrative examples: wherein each W is selected from CR 56 , C(R 56 ) 2 , NR 56 , O and S; and each Y is selected from NR 56 , O and S; and R 56 is independently hydrogen, C 1 -C 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -C 10 aryl, 5-10 membered heteroaryl,
  • These heterocycloalkyl rings may be optionally substituted with one or more groups selected from the group consisting of acyl, acylamino, acyloxy (-O-acyl or -OC(O)R 20 ), alkoxy, alkoxycarbonyl, alkoxycarbonylamino (-NR -alkoxycarbonyl or -NH-C(O)-OR 27 ), amino, substituted amino, aminocarbonyl (amido or -C(O)-NR
  • 'Nitro' refers to the radical -NO 2 .
  • 'Substituted' refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s).
  • substituted groups are substituted with one or more substituents, particularly with 1 to 3 substituents, in particular with one substituent group.
  • substituent group or groups are selected from: halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR " SO 2 R “ , -SO 2 NR R “ , -C(O)R “ , -C(O)OR “ , -OC(O)R “ , - NR '" C(O)R “ , -C(0)NR “ R “' , -NR “ R '” , -(CR '” R '” ) m OR '” , wherein, each R " is independently selected from H, Cp C 8 alkyl, -(CH 2 MC 6 -C 10 aryl), -(CH 2 ) t (5-10 membered heteroaryl), -(CH 2 MC 3 -C 10 cycloalkyl), and -(CH 2 ) t (4- 10 membered heterocycloalkyl), wherein t is an integer from
  • any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, unsubstituted C 1 -C 4 alkoxy, unsubstituted C 1 -C 4 haloalkyl, unsubstituted C 1 -C 4 hydroxyalkyl, or unsubstituted C 1 -C 4 haloalkoxy or hydroxy.
  • Each R independently represents H or C 1 -C 6 alkyl.
  • Substituted sulfanyl refers to the group -SR 61 , wherein R 61 is selected from:
  • Exemplary 'substituted sulfanyl' groups are -S-(CpC 8 alkyl) and -S-(C 3 -Ci 0 cycloalkyl), -S-
  • Exemplary 'substituted sulfinyl' groups are -S(O)-(CpC 8 alkyl) and -S(O)-(C 3 -Ci 0 cycloalkyl), -S(O)-(CH 2 MC 6 -C 10 aryl), -S(O)-(CH 2 ) t (5-10 membered heteroaryl), -S(O)-(CH 2 MC 3 -C 10 cycloalkyl), and -S(O)-(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4 and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted C 1 -C 4 alkyl, halo, unsubstituted C 1 -C 4 alkoxy, unsubstituted C 1 -C 4 haloalky
  • Exemplary 'substituted sulfonyl' groups are -S(O) 2 -(Ci-C 8 alkyl) and -S(O) 2 -(C 3 -Ci 0 cycloalkyl), -S(O) 2 -(CH 2 MC 6 -C 10 aryl), -S(O) 2 -(CH 2 ) t (5-10 membered heteroaryl), -S(O) 2 -(CH 2 MC 3 -C 10 cycloalkyl), and -S(O) 2 -(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4 and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted
  • 'Sulfo' or 'sulfonic acid' refers to a radical such as -SO 3 H.
  • 'Substituted sulfo' or 'sulfonic acid ester' refers to the group -S(O) 2 OR 82 , wherein R 82 is selected from:
  • Exemplary 'Substituted sulfo' or 'sulfonic acid ester' groups are -S(O) 2 -O-(CpC 8 alkyl) and -
  • heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
  • 'Pharmaceutically acceptable salt' refers to a salt of a compound of the invention that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts.
  • such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2- ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2- naphthalenesulfonic acid, 4-toluenesul,
  • Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like.
  • pharmaceutically acceptable cation refers to an acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like.
  • 'Pharmaceutically acceptable vehicle' refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
  • 'Prodrugs' refers to compounds, including derivatives of the compounds of the invention, which have cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention which are pharmaceutically active in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
  • 'Solvate' refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. This physical association includes hydrogen bonding.
  • solvents include water, ethanol, acetic acid and the like.
  • the compounds of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated.
  • Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • 'Solvate' encompasses both solution-phase and isolable solvates.
  • Representative solvates include hydrates, ethanolates and methanolates.
  • 'Subject' includes humans.
  • the terms 'human', 'patient' and 'subject' are used interchangeably herein.
  • 'Therapeutically effective amount means the amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease.
  • the "therapeutically effective amount” can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • 'Preventing' or 'prevention' refers to a reduction in risk of acquiring or developing a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to a disease-causing agent, or predisposed to the disease in advance of disease onset.
  • the term 'prophylaxis' is related to 'prevention', and refers to a measure or procedure the purpose of which is to prevent, rather than to treat or cure a disease.
  • Non-limiting examples of prophylactic measures may include the administration of vaccines; the administration of low molecular weight heparin to hospital patients at risk for thrombosis due, for example, to immobilization; and the administration of an antimalarial agent such as chloroquine, in advance of a visit to a geographical region where malaria is endemic or the risk of contracting malaria is high.
  • an antimalarial agent such as chloroquine
  • 'Treating' or 'treatment' of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting the disease or reducing the manifestation, extent or severity of at least one of the clinical symptoms thereof).
  • 'treating' or 'treatment' refers to ameliorating at least one physical parameter, which may not be discernible by the subject.
  • 'treating' or 'treatment' refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • "treating" or "treatment” relates to slowing the progression of the disease.
  • condition(s) involving inflammation' refers to the group of conditions including, rheumatoid arthritis, osteoarthritis, juvenile idiopathic arthritis, psoriasis, allergic airway disease (e.g. asthma, rhinitis), inflammatory bowel diseases (e.g. Crohn's disease, colitis), endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), and related diseases involving cartilage, such as that of the joints.
  • the term refers to rheumatoid arthritis, osteoarthritis, allergic airway disease (e.g. asthma) and inflammatory bowel diseases.
  • obstructive airways disease including conditions such as COPD, asthma (e.g intrinsic asthma, extrinsic asthma, dust asthma, infantily asthma) particularly chronic or inveterate asthma (for example late asthma and airway hyperreponsiveness), bronchitis, including bronchial asthma, systemic lupus erythematosus (SLE), multiple sclerosis, type I diabetes mellitus and complications associated therewith, atopic eczema (atopic dermatitis), contact dermatitis and further eczematous dermatitis, inflammatory bowel disease (e.g.
  • COPD chronic or inveterate asthma
  • bronchitis including bronchial asthma, systemic lupus erythematosus (SLE), multiple sclerosis, type I diabetes mellitus and complications associated therewith, atopic eczema (atopic dermatitis), contact dermatitis and further eczematous dermatitis, inflammatory bowel disease (
  • the term 'transplantation rejection' refers to the acute or chronic rejection of cells, tissue or solid organ allo- or xenografts of e.g. pancreatic islets, stem cells, bone marrow, skin, muscle, corneal tissue, neuronal tissue, heart, lung, combined heart-lung, kidney, liver, bowel, pancreas, trachea or oesophagus, or graft-versus-host diseases.
  • the term 'proliferative disease(s)' refers to conditions such as cancer (e.g. uterine leiomyosarcoma or prostate cancer), myeloproliferative disorders (e.g. polycythemia vera, essential thrombocytosis and myelofibrosis), leukemia (e.g. acute myeloid leukaemia and acute lymphoblastic leukemia), multiple myeloma, psoriasis, restenosis, sclerodermitis or fibrosis.
  • cancer e.g. uterine leiomyosarcoma or prostate cancer
  • myeloproliferative disorders e.g. polycythemia vera, essential thrombocytosis and myelofibrosis
  • leukemia e.g. acute myeloid leukaemia and acute lymphoblastic leukemia
  • multiple myeloma psoriasis
  • restenosis sclerodermitis or fibro
  • the term 'cancer' refers to a malignant or benign growth of cells in skin or in body organs, for example but without limitation, breast, prostate, lung, kidney, pancreas, stomach or bowel.
  • a cancer tends to infiltrate into adjacent tissue and spread (metastasise) to distant organs, for example to bone, liver, lung or the brain.
  • cancer includes both metastatic rumour cell types, such as but not limited to, melanoma, lymphoma, leukaemia, fibrosarcoma, rhabdomyosarcoma, and mastocytoma and types of tissue carcinoma, such as but not limited to, colorectal cancer, prostate cancer, small cell lung cancer and non-small cell lung cancer, breast cancer, pancreatic cancer, bladder cancer, renal cancer, gastric cancer, glioblastoma, primary liver cancer, ovarian cancer, prostate cancer and uterine leiomyosarcoma.
  • 'leukaemia' refers to neoplastic diseases of the blood and blood forming organs. Such diseases can cause bone marrow and immune system dysfunction, which renders the host highly susceptible to infection and bleeding.
  • leukemia refers to acute myeloid leukaemia (AML) and acute lymphoblastic leukemia (ALL).
  • osteoarthritis psoriatic arthritis, juvenile rheumatoid arthritis, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome or costal chondritis, fibromyalgia, osteochondritis, neurogenic or neuropathic arthritis, arthropathy, endemic forms of arthritis like osteoarthritis deformans endemica, Mseleni disease and Handigodu disease; degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma and ankylosing spondylitis.
  • the term 'congenital cartilage malformation(s)' includes conditions such as hereditary chondrolysis, chondrodysplasias and pseudochondrodysplasias, in particular, but without limitation, microtia, anotia, metaphyseal chondrodysplasia, and related disorders.
  • the term 'disease(s) associated with hypersecretion of IL6' includes conditions such as Castleman's disease, multiple myeloma, psoriasis, Kaposi's sarcoma and/or mesangial proliferative glomerulonephritis .
  • Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are particularly useful prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Particular such prodrugs are the Ci to Cg alkyl, C 2 -Cg alkenyl, aryl, C7-C 12 substituted aryl, and C7-C 12 arylalkyl esters of the compounds of the invention.
  • the term 'isotopic variant' refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such compound.
  • an 'isotopic variant' of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium ( H or D), carbon-13 ( 13 C), nitrogen-15 ( 15 N), or the like.
  • non-radioactive isotopes such as for example, deuterium ( H or D), carbon-13 ( 13 C), nitrogen-15 ( 15 N), or the like.
  • the invention may include the preparation of isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. H, and carbon- 14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • compounds may be prepared that are substituted with positron emitting isotopes, such as 11 C, 18 F, 15 O and 13 N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • PET Positron Emission Topography
  • An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively).
  • a chiral compound can exist as either individual enantiomer or as a mixture thereof.
  • a mixture containing equal proportions of the enantiomers is called a 'racemic mixture'.
  • 'Tautomers' refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of ⁇ electrons and an atom (usually H).
  • enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base.
  • Another example of tautomerism is the aci- and nitro- forms of phenylnitro methane, that are likewise formed by treatment with acid or base.
  • Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
  • the compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof.
  • R R
  • S S
  • the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof.
  • the methods for the determination of stereochemistry and the separation of stereoisomers are well- known in the art.
  • the present invention is based on the discovery that inhibitors of JAK are useful for the treatment of diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g.
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers , especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • diseases involving cartilage degradation, bone and/or joint degradation and/or inflammation for example osteoarthritis.
  • the present invention also provides methods for the production of these compounds, pharmaceutical compositions comprising these compounds and methods for treating diseases involving cartilage degradation, bone and/or joint degradation and/or inflammation by administering a compound of the invention.
  • the present compounds may be inhibitors of one or more members of the JAK family; specifically they may inhibit the activity of one or more of JAKl, JAK2, JAK3 and/or TYK2.
  • substituted bicycloheteroaryl compounds are disclosed according to Formula (I):
  • each CyI and Cy2 is independently selected from aryl and heteroaryl;
  • Ll is selected from a single bond, -O-, -C(O)-, -S(O) 2 -, -N(R 4a )-, -CON(R 4a )-, -SO 2 N(R 4a )-, -
  • Ll is selected from a single bond, -O-, -C(O)-, -S(O) 2 -, -N(R 4a )-, -CON(R 4a )-, -SO 2 N(R 4a )-, - N(R 4a )CO-, or - N(R 4a )SO 2 -; each R 1 is independently selected from unsubstituted C 1 -C 6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted amido, unsubstituted C 1 -C 6 alkoxy, unsubstituted amino, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C 3 -C 7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl
  • each R 1 is independently selected from C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, and halo.
  • each R 1 is independently selected from Me, CF 3 , Cl and F.
  • ml is 0.
  • R a is independently selected from H, Ci-Ce alkyl, and substituted Ci-C ⁇ alkyl.
  • R a is H.
  • each of R 2b , and R 2d is independently H, Ci-Ce alkyl, substituted Ci-C ⁇ alkyl, or halo.
  • each of R 2b , and R 2d is independently H, Me, F or Cl.
  • Ll is a single bond
  • nl is 0, and R 2c is H, Cl, F, Me, Et, OMe, OEt, O-i-Pr, CF 3 , CONH 2 , CONMe 2 , CONHMe,
  • Ll is a single bond
  • nl is O
  • R 2c is NHCOMe, or COOH.
  • Ll is CONH; nl is 2 or 3; and R 2c is NMe 2 , OMe, or NHCOMe.
  • Ll is selected from a single bond, -C(O)-, and -C0N(R 4a )-; nl is O, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted C3-C7 cycloalkyl, or substituted or unsubstituted heterocycloalkyl.
  • Ll is selected from a single bond, -C(O)-, and -C0N(R 4a )-; nl is O, 1, 2, 3, or 4; and R 2c is C 1 -C 6 alkyl.
  • Ll is selected from a single bond, -C(O)-, and -C0N(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is Me, Et, i-Pr, 1,3- dihydroxyprop-2-yl.
  • Ll is selected from a single bond, -C(O)-, - and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted C3-C7 cycloalkyl.
  • Ll is selected from a single bond, -C(O)-, - and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted cyclopropyl, substituted or unsubstituted cyclobutyl, substituted or unsubstituted cyclohexyl, or substituted or unsubstituted cyclopentyl.
  • Ll is selected from a single bond, -C(O)-, and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
  • Ll is selected from a single bond, -C(O)-, and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted Ph, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrrolyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted indolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted benzofuranyl, substituted or unsubstituted
  • Ll is selected from a single bond, -C(O)-, and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is substituted or unsubstituted 4-7 membered heterocycloalkyl.
  • Ll is selected from a single bond, -C(O)-, and -CON(R 4a )-; nl is 0, 1, 2, 3, or 4; and R 2c is piperidinyl, morpholinyl, piperazinyl, or pyrrolidinyl, each of which may be unsubstituted or substituted with C 1 -C 6 alkyl, acyl, phenyl, or OH.
  • Ll-(CH 2 ) n i-R 2c is selected from:
  • nl and R 2c are as described for Formula I.
  • Ll-(CH 2 ) n i-R 2c is selected from: wherein nl and R 2c are as described for Formula I.
  • Ll-(CH 2 ) n i-R 2c is selected from:
  • nl and R 2c are as described for Formula I.
  • R 2c is a substituted or unsubstituted N-containing 4-7 memberedheterocycloalkyl group or an N-containing substituted or unsubstituted 4-7 membered heteroaryl ring.
  • R c is: or
  • R c is pyrazolyl, pyrrolyl, imidazolyl, or triazolyl.
  • nl is 0, 1 or 2.
  • Ll-(CH 2 ) n i-R 2c is selected from:
  • Cy2 is Ph; and m2 is 0.
  • Cy2 is Ph; m2 is 1, 2 or 3; and each R 3a is independently Ci-Ce alkyl, Ci-C ⁇ haloalkyl, Ci-Ce alkoxy, or halo.
  • R 3a is independently Ci-Ce alkyl, Ci-C ⁇ haloalkyl, Ci-Ce alkoxy, or halo.
  • Cy2 is Ph; m2 is 1, 2 or 3; and each R 3a is independently Cl, F, Me, Et, OMe, OEt, O-i-Pr, CF 3 , OCF 3 , CONH 2 , CONMe 2 , CONHMe, SO 2 NH 2 , SO 2 NMe 2 , CN, NHCOMe, COOH, OH or COOEt.
  • the compound is according to Formula IVa, IVb, IVc, or IVd:
  • nl is 1, 2, or 3;
  • R 3a is Cl, F, Me, Et, OMe, OEt, O-i-Pr, CF 3 , OCF 3 , OCH 2 CN, CONH 2 , CONMe 2 , CONHMe, SO 2 NH 2 , SO 2 NMe 2 , CN, NHCOMe, COOH, OH or COOEt; and
  • R 2c is substituted or unsubstituted C 3 -C 7 heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R c is substituted or unsubstituted pyrrolidinyl, substituted or unsubstituted piperidinyl, substituted or unsubstituted piperazinyl, or substituted or unsubstituted morpholinyl.
  • R c is substituted or unsubstituted pyrazolyl, substituted or unsubstituted pyrrolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted indolyl, or substituted or unsubstituted indazolyl.
  • R 3a is Cl, F, Me, Et, OMe, OEt, O-i-Pr, O-i-Pr, CF 3 , OCF 3 , SO 2 NH 2 , SO 2 NMe 2 , or CN.
  • IVb, IVc or IVd, R 3a is Cl, F, Me, or OMe.
  • IVb, IVc or IVd, R 3a is OMe.
  • the compound of the invention is not an isotopic variant.
  • the compound is selected from the compounds listed in Table 1.
  • a compound of the invention according to any one of the embodiments herein described is present as the free base.
  • a compound of the invention according to any one of the embodiments herein described is a pharmaceutically acceptable salt.
  • a compound of the invention according to any one of the embodiments herein described is a solvate.
  • a compound of the invention according to any one of the embodiments herein described is a solvate of a pharmaceutically acceptable salt.
  • the present invention provides prodrugs and derivatives of the compounds according to the formulae above.
  • Prodrugs are derivatives of the compounds of the invention, which have metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutically active, in vivo.
  • Such examples include, but are not limited to, choline ester derivatives and the like, N-alkylmorpholine esters and the like.
  • Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Particularly useful are the Ci to Cg alkyl, C 2 -Cg alkenyl, aryl, C7-C 12 substituted aryl, and C7-C 12 arylalkyl esters of the compounds of the invention.
  • the compounds of this invention are typically administered in the form of a pharmaceutical composition.
  • Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • the compounds of this invention are administered in a pharmaceutically effective amount.
  • the amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound -administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal.
  • the compounds of this invention are preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration
  • the compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient, vehicle or carrier.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the furansulfonic acid compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like.
  • Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art.
  • the active compound in such compositions is typically a minor component, often being from about 0.05 to 10% by weight with the remainder being the injectable carrier and the like.
  • Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the active ingredients When formulated as a ointment, the active ingredients will typically be combined with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base.
  • Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
  • the compounds of this invention can also be administered by a transdermal device.
  • transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
  • the compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in Remington's Pharmaceutical Sciences.
  • a compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 240-270 mg tablets (80-90 mg of active amide compound per tablet) in a tablet press. Formulation 2 - Capsules
  • a compound of the invention may be admixed as a dry powder with a starch diluent in an approximate 1 :1 weight ratio. The mixture is filled into 250 mg capsules (125 mg of active amide compound per capsule).
  • a compound of the invention (125 mg), may be admixed with sucrose (1.75 g) and xanthan gum (4 mg) and the resultant mixture may be blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose (11 :89, 50 mg) in water.
  • Sodium benzoate (10 mg) flavor, and color are diluted with water and added with stirring. Sufficient water may then be added with stirring. Sufficient water is then added to produce a total volume of 5 mL.
  • a compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 450-900 mg tablets (150-300 mg of active amide compound) in a tablet press.
  • a compound of the invention may be dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/mL.
  • Stearyl alcohol (250 g) and a white petrolatum (250 g) may be melted at about 75°C and then a mixture of a compound of the invention (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) is added and the resulting mixture is stirred until it congeals.
  • the present compounds may be used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of JAK.
  • a compound of the invention and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis; and/or conditions involving inflammation or immune responses, such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g.
  • Inhibitors of JAK can also find application in the treatment of proliferative diseases.
  • the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer).
  • the conditions are selected from inflammatory conditions, conditions related to cartilage and/or joint degradation in mammals including humans.
  • the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating proliferative disorders in mammals, including humans.
  • the compound of the invention and pharmaceutical compositions thereof find use as therapeutics for preventing and/or treating cancer in mammals including humans.
  • this invention provides methods of treating a mammal susceptible to or afflicted with condition involving an immune response or an autoimmune disease.
  • the methods comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compound of the invention herein described.
  • the autoimmune disease is selected from COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and inflammatory bowel disease.
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of a condition involving an autoimmune response or an autoimmune disease.
  • the autoimmune disease is selected from COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and inflammatory bowel disease.
  • this invention provides a method of treatment, prevention or prophylaxis in a mammal susceptible to or afflicted with diseases involving impairment of cartilage turnover (e.g. a condition associated with, or diseases involving the anabolic stimulation of chondrocytes), for example, osteoarthritis, psoriatic arthritis, juvenile rheumatoid arthritis, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome or costal chondritis, fibromyalgia, osteochondritis, neurogenic or neuropathic arthritis, arthropathy, endemic forms of arthritis like osteoarthritis deformans endemica, Mseleni disease and Handigodu disease; degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma and ankylosing spondylitis, which method comprises administering a condition associated with, or diseases involving the an
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of diseases involving impairment of cartilage turnover (e.g. a condition associated with, or diseases involving the anabolic stimulation of chondrocytes), for example, osteoarthritis, psoriatic arthritis, juvenile rheumatoid arthritis, gouty arthritis, septic or infectious arthritis, reactive arthritis, reflex sympathetic dystrophy, algodystrophy, Tietze syndrome or costal chondritis, fibromyalgia, osteochondritis, neurogenic or neuropathic arthritis, arthropathy, endemic forms of arthritis like osteoarthritis deformans endemica, Mseleni disease and Handigodu disease; degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma and ankylosing spondylitis.
  • diseases involving impairment of cartilage turnover e.g. a condition associated with, or diseases
  • the present invention also provides a method of treatment of congenital cartilage malformations, including hereditary chondrolysis, chondrodysplasias and pseudochondrodysplasias, in particular, but without limitation, microtia, anotia, metaphyseal chondrodysplasia, and related disorders, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of congenital cartilage malformations, including hereditary chondrolysis, chondrodysplasias and pseudochondrodysplasias, in particular, but without limitation, microtia, anotia, metaphyseal chondrodysplasia, and related disorders.
  • this invention provides a method of treating a mammal susceptible to or afflicted with a condition involving inflammation, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • this invention provides methods of treating a mammal susceptible to or afflicted with diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, allergic airway disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g.
  • the method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • the condition involving inflammation is selected from rheumatoid arthritis, osteoarthritis, allergic airway disease (e.g. asthma) and inflammatory bowel diseases.
  • the methods comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds herein described.
  • this invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of a condition involving inflammation.
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of diseases and disorders which are mediated by or result in inflammation such as, for example rheumatoid arthritis and osteoarthritis, allergic airway disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis, inflammatory bowel diseases, endotoxin- driven disease states (e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure), and related diseases involving cartilage, such as that of the joints.
  • the condition involving inflammation is selected from rheumatoid arthritis, osteoarthritis, allergic airway disease (e.g. asthma) and inflammatory bowel diseases.
  • this invention provides methods of treating a mammal susceptible to or afflicted with a proliferative disease, in particular cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), leukemia (e.g. AML or ALL), multiple myeloma and/or psoriasis, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • cancer e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer
  • leukemia e.g. AML or ALL
  • this invention provides methods of treating a mammal susceptible to or afflicted with cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer) and/or leukemias, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • cancer e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer
  • leukemias e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of a proliferative disease, in particular cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), leukemia (e.g. AML or ALL), multiple myeloma and/or psoriasis.
  • cancer e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer
  • leukemia e.g. AML or ALL
  • multiple myeloma and/or psoriasis e.g. AML or ALL
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of cancer (e.g solid tumors such as uterine leiomyosarcoma or prostate cancer) and/or leukemias.
  • cancer e.g solid tumors such as uterine leiomyosarcoma or prostate cancer
  • leukemias e.g solid tumors such as uterine leiomyosarcoma or prostate cancer
  • this invention provides methods of treating a mammal susceptible to or afflicted with diseases associated with hypersecretion of IL6, in particular Castleman's disease or mesangial proliferative glomerulonephritis, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of diseases associated with hypersecretion of IL6, in particular
  • this invention provides methods of treating a mammal susceptible to or afflicted with transplantation rejection, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • the invention provides methods of treating organ transplant rejection, which method comprises administering a therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions herein described.
  • the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of transplantation rejection.
  • the invention provides methods of treating organ transplant rejection.
  • the present compounds for use as a pharmaceutical especially in the treatment or prevention of the aforementioned conditions and diseases. Also provided herein is the use of the present compounds in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases.
  • a particular regimen of the present method comprises the administration to a subject in suffering from a disease involving inflammation, of an effective amount of a compound of the invention for a period of time sufficient to reduce the level of inflammation in the patient, and preferably terminate, the processes responsible for said inflammation.
  • a special embodiment of the method comprises administering of an effective amount of a compound of the invention to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, inflammation in the joints of said patient, and preferably terminate, the processes responsible for said inflammation.
  • a further particular regimen of the present method comprises the administration to a subject in suffering from a disease condition characterized by cartilage or joint degradation (e.g. osteoarthritis) of an effective amount of a compound of the invention for a period of time sufficient to reduce and preferably terminate, the self-perpetuating processes responsible for said degradation.
  • a special embodiment of the method comprises administering of an effective amount of a compound of the invention to a subject patient suffering from or susceptible to the development of osteoarthritis, for a period of time sufficient to reduce or prevent, respectively, cartilage degradation in the joints of said patient, and preferably terminate, the self- perpetuating processes responsible for said degradation.
  • said compounds exhibit cartilage anabolic and/or anti-catabolic properties.
  • Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours.
  • a preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels.
  • the maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
  • each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with particular doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.
  • Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
  • a compound of the invention When used to prevent the onset of an inflammatory condition, a compound of the invention will be administered to a patient at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above. Patients at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition.
  • the compounds of the invention can be administered as the sole active agent or they can be administered in combination with other agents, including other compounds that demonstrate the same or a similar therapeutic activity, and that are determined to safe and efficacious for such combined administration. In a specific embodiment, co-administration of two (or more) agents allows for significantly lower doses of each to be used, thereby reducing the side effects seen.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of a disease involving inflammation;
  • agents include, but are not limited to, immunoregulatory agents e.g. azathioprine, corticosteroids (e.g. prednisolone or dexamethasone), cyclophosphamide, cyclosporin A, tacrolimus, Mycophenolate Mofetil, muromonab-CD3 (OKT3, e.g. Orthocolone®), ATG, aspirin, acetaminophen, ibuprofen, naproxen, and piroxicam.
  • immunoregulatory agents e.g. azathioprine, corticosteroids (e.g. prednisolone or dexamethasone), cyclophosphamide, cyclosporin A, tacrolimus, Mycophenolate Mofetil, muromonab-CD3 (OKT3, e
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of arthritis (e.g. rheumatoid arthritis); particular agents include but are not limited to analgesics, non-steroidal anti-inflammatory drugs (NSAIDS), steroids, synthetic DMARDS (for example but without limitation methotrexate, leflunomide, sulfasalazine, auranofm, sodium aurothiomalate, penicillamine, chloroquine, hydroxychloroquine, azathioprine, and ciclosporin), and biological DMARDS (for example but without limitation Infliximab, Etanercept, Adalimumab, Rituximab, and Abatacept).
  • NSAIDS non-steroidal anti-inflammatory drugs
  • DMARDS for example but without limitation methotrexate, leflunomide, sulfasalazine, auranofm, sodium aurothiomalate, penicillamine, chlor
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of proliferative disorders; particular agents include but are not limited to: methotrexate, leukovorin, adriamycin, prenisone, bleomycin, cyclophosphamide, 5-fluorouracil, paclitaxel, docetaxel, vincristine, vinblastine, vinorelbine, doxorubicin, tamoxifen, toremifene, megestrol acetate, anastrozole, goserelin, anti- HER2 monoclonal antibody (e.g.
  • a compound of the invention may be administered in combination with other therapies including, but not limited to, radiotherapy or surgery.
  • the proliferative disorder is selected from cancer, myeloproliferative disease or leukaemia.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of autoimmune diseases
  • agents include but are not limited to: glucocorticoids, cytostatic agents (e.g. purine analogs), alkylating agents, (e.g nitrogen mustards (cyclophosphamide), nitrosoureas, platinum compounds, and others), antimetabolites (e.g. methotrexate, azathioprine and mercaptopurine), cytotoxic antibiotics (e.g.
  • dactinomycin anthracyclines mitomycin C, bleomycin, and mithramycin
  • antibodies e.g., anti-CD20, anti-CD25 or anti-CD3 (OTK3) monoclonal antibodies, Atgam® and Thymoglobuline®
  • cyclosporin tacrolimus, rapamycin (sirolimus), interferons (e.g. IFN- ⁇ ), TNF binding proteins (e.g. infliximab (Remicade), etanercept (Enbrel), or adalimumab (Humira)), mycophenolate, Fingolimod, Myriocin.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of transplantation rejection
  • therapeutic agents include but are not limited to: calcineurin inhibitors (e.g. cyclosporin or tacrolimus (FK506)), mTOR inhibitors (e.g. sirolimus, everolimus), anti-proliferatives (e.g. azathioprine, mycophenolic acid), corticosteroids (e.g. prednisolone, hydrocortisone), Antibodies (e.g. monoclonal anti-IL-2R ⁇ receptor antibodies, basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of asthma and/or rhinitis and/or COPD, particular agents include but are not limited to: beta 2 -adrenoceptor agonists (e.g. salbutamol, levalbuterol, terbutaline and bitolterol.), epinephrine (inhaled or tablets), anticholinergics (e.g. ipratropium bromide), glucocorticoids (oral or inhaled) Long-acting ⁇ 2 -agonists (e.g.
  • salmeterol, formoterol, bambuterol, and sustained-release oral albuterol combinations of inhaled steroids and long-acting bronchodilators (e.g. fluticasone/salmeterol, budesonide/formoterol), leukotriene antagonists and synthesis inhibitors (e.g. montelukast, zafirlukast and zileuton), inhibitors of mediator release (e.g. cromoglycate and ketotifen), biological regulators of IgE response (e.g. omalizumab), antihistamines (e.g. ceterizine, cinnarizine, fexofenadine), vasoconstrictors (e.g. oxymethazoline, xylomethazoline, nafazoline and tramazoline).
  • bronchodilators e.g. fluticasone/salmeterol, budesonide/formoterol
  • a compound of the invention may be administered in combination with emergency therapies for asthma and/or COPD, such therapies include oxygen or heliox administration, nebulized salbutamol or terbutaline (optionally combined with an anticholinergic (e.g. ipratropium), systemic steroids (oral or intravenous, e.g. prednisone, prednisolone, methylprednisolone, dexamethasone, or hydrocortisone), intravenous salbutamol, nonspecific beta-agonists, injected or inhaled (e.g.
  • oxygen or heliox administration ebulized salbutamol or terbutaline
  • an anticholinergic e.g. ipratropium
  • systemic steroids oral or intravenous, e.g. prednisone, prednisolone, methylprednisolone, dexamethasone, or hydrocortisone
  • intravenous salbutamol e.g. predn
  • epinephrine isoetharine, isoproterenol, metaproterenol
  • anticholinergics IV or nebulized, e.g. glycopyrrolate, atropine, ipratropium
  • methylxanthines theophylline, aminophylline, bamiphylline
  • inhalation anesthetics that have a bronchodilatory effect (e.g. isoflurane, halothane, enflurane), ketamine, intravenous magnesium sulfate.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of IBD
  • agents include but are not limited to: glucocorticoids (e.g. prednisone, budesonide) synthetis disease modifying, immunomodulatory agents (e.g. methotrexate, leflunomide, sulfasalazine, mesalazine, azathioprine, 6-mercaptopurine and ciclosporin) and biological disease modifying, immunomodulatory agents (infliximab, adalimumab, rituximab, and abatacept).
  • glucocorticoids e.g. prednisone, budesonide
  • immunomodulatory agents e.g. methotrexate, leflunomide, sulfasalazine, mesalazine, azathioprine, 6-mercaptopurine and ciclosporin
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of SLE
  • particular agents include but are not limited to: Disease- modifying antirheumatic drugs (DMARDs) such as antimalarials (e.g. plaquenil, hydroxychloroquine), immunosuppressants (e.g. methotrexate and azathioprine), cyclophosphamide and mycophenolic acid; immunosuppressive drugs and analgesics, such as nonsteroidal anti-inflammatory drugs, opiates (e.g. dextropropoxyphene and co-codamol), opioids (e.g. hydrocodone, oxycodone, MS Contin, or methadone) and the fentanyl duragesic transdermal patch.
  • DMARDs Disease- modifying antirheumatic drugs
  • antimalarials e.g. plaquenil, hydroxychloroquine
  • immunosuppressants e.g.
  • a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of psoriasis
  • agents include but are not limited to: topical treatments such as bath solutions, moisturizers, medicated creams and ointments containing coal tar, dithranol (anthralin), corticosteroids like desoximetasone (Topicort), fluocinonide, vitamin D 3 analogues (for example, calcipotriol), Argan oiland retinoids (etretinate, acitretin, tazarotene), systemic treatments such as methotrexate, cyclosporine, retinoids, tioguanine, hydroxyurea, sulfasalazine, mycophenolate mofetil, azathioprine, tacrolimus, fumaric acid esters or biologies such as Amevive, Enbrel, Humira, Remicade,
  • any means of delivering two or more therapeutic- agents to the patient as part of the same treatment regime is included any means of delivering two or more therapeutic- agents to the patient as part of the same treatment regime, as will be apparent to the skilled person. Whilst the two or more agents may be administered simultaneously in a single formulation this is not essential. The agents may be administered in different formulations and at different times.
  • the compounds of the invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • a compound of the invention can be produced according to the following scheme.
  • Ar is -Cy2-(R 3a ) m2 -; and Ar'is -Cyl-Ll-(CH 2 ) n i-R 2c ; and CyI, Cy2, Ll, L2, nl, m2,
  • R 2c , and R 3a are as described herein.
  • Step a Preparation of 2-amino-5-Ar-triazolopyridine derivatives
  • An appropriate Ar substituted boronic acid derivative (2eq.) is added to a solution of 2-amino-8- bromo-triazolopyridine (Commercially available, BiofocusDPI) in dioxan/water (5: 1).
  • K 2 CO 3 (2 eq.) and PdCl 2 dppf (5%) are added to the mixture.
  • the resulting mixture is heated in a microwave oven at 140 0 C for 15-45 min or heated in an oil bath at 90 0 C for 4 to 16 h until the reaction goes to completion (monitored by LCMS). Water is added and the mixture is extracted with ethyl acetate.
  • reaction mixture is extracted with ethyl acetate and the extracts are combined, washed with water and dried over anhyd. magnesium sulfate.
  • the organic solvent is removed under high vacuum to yield the crude product.
  • the crude product is then purified by flash chromatography to give the corresponding 5-Ar-2-iodo-triazolopyridine derivative (3).
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4'- aminoacetanilide.
  • Compound 2 [00240] This compound was prepared via Method A using 4-methoxyphenylboronic acid and 3- aminopyridine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and p- tolylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4-(1H- pyrazol- 1 -yl)aniline.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4-ethyl- phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 6- morpholin-4-yl-pyridin-3-ylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4- morpholin-4-yl-phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 6-(4- methylpiperazin- 1 -yl)pyridin-3 -amine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4- pyridin-3 -ylaniline.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 6- methoxy-pyridin-3 -ylamine.
  • Step 2 [00271] 4-hydroxyphenylboronic acid (1.2eq.) was added to a solution of 4-(5-Bromo-
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4-amino- benzoic acid methyl ester.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and benzooxazol-6-ylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4-[4-(4- fluoro-phenyl)-piperazin- 1 -yl] -phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4-(4- isopropyl-piperazin- 1 -yl)-phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4- pyrazol- 1 -ylmethyl-phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenylboronic acid and 4- imidazol- 1 -ylmethyl-phenylamine.
  • Step b
  • This compound was prepared using the same method as described for Compound 39 using 2- pyrrolidin- 1 -yl-ethylamine.
  • Step a (5-Bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-(4-nitro-phenyl)-amine
  • Step c N-[4-(5-Bromo-[ 1 ,2,4] 'triazolo[ 1 ,5-aj 'pyridin-2-ylamino) -phenyl] '-acetamide
  • Step b N[4-(4,4,5,5,-tetramethyl-l,3,2-dioxaborolan-2-yl)-methanesulfonamide (2eq.) was added to a solution of 4-(5-Bromo-[l,2,4]triazolo[l,5-a]pyridin-2-ylamino)-benzoic acid ethyl ester in 1 ,4-dioxane/water (5: 1) (or EtOH). K 2 CO 3 (2 eq.) and PdCl 2 dppf (cat.) were added to the mixture.
  • This compound was prepared using the same method as described for Compound 39 using 2- pyrrolidin- 1 -yl-ethylamine.
  • N(6-chloro-pyridin-3-yl)-[5-(4-methoxy-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amine prepared by method A (leq) in dioxan/water (5:1).
  • K 2 CO3 (2 eq.) and PdC ⁇ dppf (cat.) were added to the mixture.
  • the reaction went to completion (monitored by LCMS). Water was added and the mixture was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous MgS ⁇ 4 and evaporated in vacuo to yield the crude product.
  • the crude product was then purified by preparative HPLC.
  • This compound was prepared via Method A using 4-methoxyphenyl boronic acid and 4-fluoro aniline.
  • This compound was prepared via Method A using 4-methoxyphenyl boronic acid and 3-fluoro- phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenyl boronic acid l-(4-Amino- phenyl)-ethanone.
  • This compound was prepared via Method A using 4-methoxyphenyl boronic acid and 3,4,5- trimethoxy-phenylamine.
  • This compound was prepared via Method A using 4-methoxyphenyl boronic acid and 2-(4- amino-phenyl)-propan-2-ol.
  • the reaction is stirred at 100 0 C for 16 hrs.
  • the crude mixture is purified by preparative HPLC.
  • the compound can be made via Method C using cyclopropylamine and 4-formylphenylboronic acid followed by this step:
  • Example 1 - in vitro assays Example 1.1 JAKl inhibition assay
  • Recombinant human JAKl catalytic domain (amino acids 850-1154; catalog number 08-144) was purchased from Carna Biosciences. 10 ng of JAKl was incubated with 12.5 ⁇ g polyGT substrate (Sigma catalog number P0275) in kinase reaction buffer (15 mM Tris-HCl pH 7.5, 1 mM DTT, 0.01% Tween-20, 10 mM MgCl 2 , 2 ⁇ M non-radioactive ATP, 0.25 ⁇ Ci 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5 ⁇ L containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 ⁇ L, in a polypropylene 96-well plate (Greiner, V-bottom).
  • kinase reaction buffer 15 mM Tris-HCl pH 7.5, 1 mM DTT, 0.01% Tween-20, 10 mM MgCl 2
  • Percentage inhibition ((cpm determined for sample with test compound present - cpm determined for sample with positive control inhibitor) divided by (cpm determined in the presence of vehicle - cpm determined for sample with positive control inhibitor)) * 100%.
  • Recombinant human JAK2 catalytic domain (amino acids 808-1132; catalog number PV4210) was purchased from Invitrogen. 0.025mU of JAK2 was incubated with 2.5 ⁇ g polyGT substrate (Sigma catalog number P0275) in kinase reaction buffer (5 mM MOPS pH 7.5, 9 mM MgAc, 0.3mM EDTA, 0.06% Brij and 0.6 mM DTT, 1 ⁇ M non-radioactive ATP, 0.25 ⁇ Ci 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5 ⁇ L containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 ⁇ L, in a polypropylene 96-well plate (Greiner, V-bottom).
  • DMSO 1% final concentration
  • Percentage inhibition ((cpm determined for sample with test compound present - cpm determined for sample with positive control inhibitor) divided by (cpm determined in the presence of vehicle - cpm determined for sample with positive control inhibitor)) * 100% .
  • Dose dilution series were prepared for the compounds enabling the testing of dose-response effects in the JAK2 assay and the calculation of the IC 5 O for each compound.
  • Each compound was routinely tested at concentration of 20 ⁇ M followed by a 1/3 serial dilution, 8 points (20 ⁇ M - 6.67 ⁇ M - 2.22 ⁇ M - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO.
  • potency of compound series increased, more dilutions were prepared and/or the top concentration was lowered (e.g. 5 ⁇ M, 1 ⁇ M).
  • Recombinant human JAK3 catalytic domain (amino acids 781-1124; catalog number PV3855) was purchased from Invitrogen. 0.025mU of JAK3 was incubated with 2.5 ⁇ g polyGT substrate (Sigma catalog number P0275) in kinase reaction buffer (25 mM Tris pH 7.5, 0.5 mM EGTA, 0.5 mM Na3VO4, 5 mM b- glycerolphosphate, 0.01% Triton X-100, 1 ⁇ M non-radioactive ATP, 0.25 ⁇ Ci 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5 ⁇ L containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 ⁇ L, in a polypropylene 96-well plate (Greiner, V-bottom).
  • DMSO 1% final concentration
  • Recombinant human TYK2 catalytic domain (amino acids 871-1187; catalog number 08-147) was purchased from Carna biosciences. 5 ng of TYK2 was incubated with 12.5 ⁇ g polyGT substrate (Sigma catalog number P0275) in kinase reaction buffer (25 mM Hepes pH 7.5, 100 mM NaCl, 0.2 mM Na3VO4, 0.1% NP-40, 0.1 ⁇ M non-radioactive ATP, 0.125 ⁇ Ci 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5 ⁇ L containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 ⁇ L, in a polypropylene 96-well plate (Greiner, V-bottom).
  • kinase reaction buffer 25 mM Hepes pH 7.5, 100 mM NaCl, 0.2 mM Na3VO4, 0.1% NP-40, 0.1 ⁇ M
  • Dose dilution series were prepared for the compounds enabling the testing of dose-response effects in the TYK2 assay and the calculation of the IC 5 O for each compound.
  • Each compound was routinely tested at concentration of 20 ⁇ M followed by a 1/3 serial dilution, 8 points (20 ⁇ M - 6.67 ⁇ M - 2.22 ⁇ M - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO.
  • potency of compound series increased, more dilutions were prepared and/or the top concentration was lowered (e.g. 5 ⁇ M, 1 ⁇ M).
  • Example 2 Cellular assays
  • DMEM Dulbecco's Modified Eagle's Medium
  • Percentage inhibition ((fluorescence determined in the presence of vehicle - fluorescence determined for sample with test compound present) divided by (fluorescence determined in the presence of vehicle - fluorescence determined for sample without trigger)) * 100 %.
  • OSM and IL- l ⁇ were shown to synergistically upregulate MMP 13 levels in the human chondrosarcoma cell line SW1353.
  • the cells were seeded in 96 well plates at 15,000 cells/well in a volume of 120 ⁇ L DMEM (Invitrogen) containing 10% (v/v) FBS and 1% penicillin/streptomycin (InVitrogen) incubated at 37°C 5% CO 2 .
  • MMP 13 levels were measured in conditioned medium 48 hours after triggering.
  • MMP 13 activity was measured using an antibody capture activity assay.
  • 384 well plates (NUNC, 460518, MaxiSorb black) were coated with 35 ⁇ L of a 1.5 ⁇ g/mL anti-human MMP 13 antibody (R&D Systems, MAB511) solution for 24 hours at 4°C.
  • Percentage inhibition ((fluorescence determined in the presence of vehicle - fluorescence determined for sample with test compound present) divided by (fluorescence determined in the presence of vehicle - fluorescence determined for sample without trigger)) * 100 %.
  • PBL Human peripheral blood lymphocytes
  • the PBL are first stimulated for 72 hrs with PHA to induce IL-2 receptor, fasted for 24 hrs to stop cell proliferation followed by IL-2 stimulation for another 72 hrs (including 24hr BrdU labeling).
  • Cells are preincubated with test compounds 1 hr before IL-2 addition.
  • Cells are cultured in RPMI 1640 containing 10% (v/v) FBS.
  • Example 3 In vivo models
  • CFA Completed Freund's adjuvant
  • IFA incomplete Freund's adjuvant
  • Bovine collagen type II CII
  • LPS lipopolysaccharide
  • Enbrel obtained from Chondrex (Isle d'Abeau, France); Sigma (P4252, L'Isle d'Abeau, France), Whyett (25mg injectable syringe, France) Acros Organics (Palo Alto, CA), respectively. All other reagents used were of reagent grade and all solvents were of analytical grade.
  • DBA/IJ mice male, 7 weeks old were obtained from Centre d'Elevage et de Reproduction JANVIER (CERJ) (Laval, France). Rats and mice were kept on a 12 hours light/dark cycle (0700 - 1900). The temperature was maintained at 22°C, and food and water were provided ad libitum.
  • mice 0.1 mL of the emulsion was injected intradermally at the base of the tail of each mouse on day 1, a second booster intradermal injection (CII solution at 1 mg/mL in CFA 0.1 mL saline) was performed on day 21.
  • This immunization method was modified from published methods (David D Brand Kary A Latham, & Edward F Rosloniec. Collagen-induced arthritis. Nature Methods 2 (5): 1269-1275, 2007).
  • Rat 0.2 mL of the emulsion was injected intradermally at the base of the tail of each rat on day
  • test compounds were tested in the rat or mouse CIA model.
  • mice were randomly divided into equal groups and each group contained 10 animals. All rats were immunized on day 1 and boosted on day 9. All mice were immunized on day 1 and boosted on day 21. Therapeutic dosing lasted from day 16 to day 30.
  • the negative control group was treated with vehicle (MC 0,5%) and the positive control group with Enbrel (10 mg/kg, 3x week., s.c).
  • Enbrel 10 mg/kg, 3x week., s.c
  • a compound of interest was typically tested at 3 doses, e.g. 3, 10, 30 mg/kg, p.o.
  • X-ray photos were taken of the hind paws of each individual animal. A random blind identity number was assigned to each of the photos, and the severity of bone erosion was ranked by two independent scorers with the radiological Larsen's score system as follows: 0- normal with intact bony outlines and normal joint space; 1- slight abnormality with any one or two of the exterior metatarsal bones showing slight bone erosion; 2-def ⁇ nite early abnormality with any three to five of the exterior metatarsal bones showing bone erosion; 3 -medium destructive abnormality with all the exterior metatarsal bones as well as any one or two of the interior metatarsal bones showing definite bone erosions; 4-severe destructive abnormality with all the metatarsal bones showing definite bone erosion and at least one of the inner metatarsal joints completely eroded leaving some bony joint outlines partly preserved; 5-mutilating abnormality without bony outlines.
  • This scoring system is a modification from Salvemini et al., 2001; Bush et al., 2002; Sims et
  • mice were fixed in 10% phosphate-buffered formalin (pH 7.4), decalcified with rapid bone decalcif ⁇ ant for fine histology (Laboratories Eurobio) and embedded in paraffin.
  • pH 7.4 10% phosphate-buffered formalin
  • H&E hematoxylin and eosin
  • Histologic examinations for synovial inflammation and bone and cartilage damage were performed double blind.
  • four parameters were assessed using a four-point scale. The parameters were cell infiltration, pannus severity, cartilage erosion and bone erosion. Scoring was performed as follows: 1 -normal, 2-mild, 3-moderate, 4-marked. These four scores were summed together and represented as an additional score, namely the 'RA total score'.
  • Bone degradation observed in RA occurs especially at the cortical bone and can be revealed by ⁇ CT analysis (Sims NA et al, 2004; Oste L et al, ECTC Montreal 2007). After scanning and 3D volume reconstruction of the calcaneus bone, bone degradation was measured as the number of discrete objects present per slide, isolated in silico perpendicular to the longitudinal axis of the bone. The more the bone that was degraded, the more discrete objects that were measured. 1000 slices, evenly distributed along the calcaneus
  • LPS lipopolysaccharide
  • TNF-alpha TNF-alpha
  • mice Six BALB/cJ female mice (20 g) per group were treated at the intended dosing once, po. Thirty minutes later, LPS (15 ⁇ g/kg; E. CoIi serotype 0111 :B4) was injected ip. Ninety minutes later, mice were euthanized and blood was collected. Circulating TNF alpha levels were determined using commercially available ELISA kits. Dexamethasone (5 ⁇ g/kg) was used as a reference anti-inflammatory compound.
  • Selected compounds are tested at one or multiple doses, e.g. 3 and/or 10 and/or 30 mg/kg, po.
  • the MAB model allows a rapid assessment of the modulation of an RA-like inflammatory response by therapeutics (Kachigian LM. Nature Protocols (2006) 2512-2516: Collagen antibody- induced arthritis).
  • DBA/J mice are injected i.v. with a cocktail of mAbs directed against collagen II.
  • compound treatment is initiated (vehicle: 10% (v/v) HP ⁇ CD).
  • mice receive an i.p. LPS injection (50 ⁇ g/mouse), resulting in a fast onset of inflammation.
  • Compound treatment is continued until 10 days after the mAb injection.
  • Inflammation is read by measuring paw swelling and recording the clinical score of each paw.
  • the cumulative clinical arthritis score of four limbs is presented to show the severity of inflammation.
  • a scoring system is applied to each limb using a scale of 0-4, with 4 being the most severe inflammation.
  • a solution of 1 mg/mL of the test compound is prepared in a 0.2M phosphate buffer pH7.4 or a
  • DMSO stock solution diluted factor 2 in DMSO (5000 ⁇ M) and then further diluted in DMSO up to 19.5 ⁇ M.
  • 3 ⁇ L of the dilution series as from 5000 ⁇ M is then transferred to a 97 ⁇ L acetonitrile-buffer mixture (50/50).
  • the final concentration range is 2.5 to 150 ⁇ M.
  • the plate is sealed with sealing mats (MA96RD-04S, www.kinesis.co.uk) and samples are measured at room temperature on LCMS (ZQ 1525 from Waters) under optimized conditions using
  • DMSO DMSO
  • the dilution series is transferred to a 96 NUNC Maxisorb plate F-bottom (Cat no. 442404) and 0.2M phosphate buffer pH7.4 or 0. IM citrate buffer pH3.0 at room temperature is added.
  • the final concentration ranged from 200 ⁇ M to 2.5 ⁇ M in 5 equal dilution steps.
  • DMSO concentration did not exceed 2%.
  • 200 ⁇ M Pyrene is added to the corner points of each 96 well plate and serves as a reference point for calibration of Z-axis on the microscope.
  • the assay plates are sealed and incubated for 1 hour at 37°C while shaking at 230rpm.
  • the plates are then scanned under a white light microscope, yielding individual pictures of the precipitate per concentration.
  • the precipitate is analyzed and converted into a number which is plotted onto a graph.
  • the first concentration at which the compound appears completely dissolved is the concentration reported, however the true concentration lies somewhere between this concentration and one dilution step higher.
  • a 1OmM stock solution of the compound in DMSO is diluted with a factor 5 in DMSO. This solution is further diluted in freshly thawed human, rat, mouse or dog plasma (BioReclamation INC) with a final concentration of lO ⁇ M and final DMSO concentration of 0.5% (5.5 ⁇ l in 1094.5 ⁇ l plasma in a PP-
  • a Pierce Red Device plate with inserts (ThermoScientific, Cat no. 89809) is prepared and filled with 750 ⁇ L PBS in the buffer chamber and 500 ⁇ L of the spiked plasma in the plasma chamber. The plate is incubated for 4 hours at 37°C while shaking at 230rpm. After incubation, 120 ⁇ L of both chambers is transferred to 360 ⁇ L acetonitrile in a 96-well round bottom, PP deep-well plates (Nunc, Cat no. 278743) and sealed with an aluminum foil lid. The samples are mixed and placed on ice for 30min. This plate is then centrifuged 30 min at 1200rcf at 4°C and the supernatant is transferred to a 96 v-bottom PP plate (Greiner,
  • the plate is sealed with sealing mats (MA96RD-04S) of www.kinesis.co.uk and samples are measured at room temperature on LCMS (ZQ 1525 from Waters) under optimized conditions using
  • Peak area from the compound in the buffer chamber and the plasma chamber are considered to be 100% compound.
  • the percentage bound to plasma is derived from these results and was reported to the
  • Pulse v8.77 software (HEKA). Series resistance is typically less than 10 M ⁇ and compensated by greater than 60%, recordings are not leak subtracted. Electrodes are manufactured from GC 150TF pipette glass (Harvard). [00487] The external bathing solution contains: 135 mM NaCl, 5 mM KCl, 1.8 mM CaCl 2 , 5 mM
  • Glucose 10 mM HEPES, pH 7.4.
  • the internal patch pipette solution contains: 10OmM Kgluconate, 20 mM KCl, ImM CaCl 2 , 1 mM MgCl 2 , 5mM Na 2 ATP, 2mM Glutathione, 11 mM EGTA, 10 mM HEPES, pH 7.2.
  • Drugs are perfused using a Biologic MEV-9/EVH-9 rapid perfusion system.
  • a 1OmM stock solution of compound in DMSO was diluted 1000 fold in a 182 mM phosphate buffer pH7.4 in a 96 deep well plate (Greiner, Cat no.780285) and pre-incubated at 37°C.
  • G6PDH Glucose-6-phophate-dehydrogenase
  • liver microsomes (Xenotech) of a species of interest
  • Caco-2 assays were performed as described below. Caco-2 cells were obtained from European Collection of Cell Cultures (ECACC, cat 86010202) and used after a 21 day cell culture in 24- well Transwell plates (Fisher TKT-545-020B).
  • Test and reference compounds (propranolol and rhodaminel23 or vinblastine, all purchased from Sigma) were prepared in Hanks' Balanced Salt Solution containing 25 mM HEPES (pH7.4) and added to either the apical (125 ⁇ L) or basolateral (600 ⁇ L) chambers of the Transwell plate assembly at a concentration of 10 ⁇ M with a final DMSO concentration of 0.25%.
  • Lucifer Yellow (Sigma) was added to the donor buffer in all wells to assess integrity of the cell layers by monitoring Lucifer Yellow permeation. As Lucifer Yellow (LY) cannot freely permeate lipophilic barriers, a high degree of LY transport indicates poor integrity of the cell layer. [00510] After a 1 hour incubation at 37°C while shaking at an orbital shaker at 150rpm, 70 ⁇ L aliquots were taken from both apical (A) and basal (B) chambers and added to lOO ⁇ Ll 50:50 acetonitrile:water solution containing analytical internal standard (0.5 ⁇ M carbamazepine) in a 96 well plate.
  • A apical
  • B basal
  • Lucifer yellow was measured with a Spectramax Gemini XS (Ex 426nm and Em 538nm) in a clean 96 well plate containing 150 ⁇ L of liquid from basolateral and apical side.
  • V chamber volume
  • T mc incubation time.
  • Surface area 0.33cm 2
  • Rhodamine 123 or Vinblastine P app (A>B) value ⁇ 5 (xlO 6 cm/s) with Efflux ratio >5.
  • Compounds are formulated in PEG200/physiological saline or PEG400/DMSO/physiological saline mixtures for the intravenous route and in 0.5% methylcellulose or 10-30% hydroxylpropyl- ⁇ - cyclodextrine pH3 or pH7.4 for the oral route.
  • Test compounds are orally dosed as a single esophageal gavage at 5-10 mg/kg and intravenously dosed as a bolus via the caudal vein at 1 mg/kg.
  • Each group consists of 3 rats.
  • Blood samples are collected either via the jugular vein using cannulated rats or at the retro-orbital sinus with lithium heparin as anti-coagulant at the time points in the following range: 0.05 to 8 hours (intravenous route), and 0.25 to 6 or 24 hours (oral route).
  • Whole blood samples are centrifuged at 5000 rpm for 10 min and the resulting plasma samples are stored at -20 0 C pending analysis.
  • Plasma concentrations of each test compound are determined by an LC-MS/MS method in which the mass spectrometer is operated in positive electrospray mode. 4.7.3 Determination of pharmacokinetic parameters
  • a 7-day oral toxicity study with test compounds was performed in Sprague-Dawley male rats to assess their toxic potential and toxicokinetics, at daily doses of 100, 300 and 500 mg/kg/day, by gavage, at the constant dosage- volume of 5 mL/kg/day.
  • test compounds were formulated in 30% (v/v) HP ⁇ CD in purified water. Each group included 5 principal male rats as well as 3 satellite animals for toxicokinetics. A fourth group was given 30%
  • Interleukin (IL)-4 and IL- 13 up-regulate monocyte chemoattractant protein- 1 expression in human bronchial epithelial cells: involvement of p38 mitogen-activated protein kinase, extracellular signal- regulated kinase 1/2 and Janus kinase-2 but not c-Jun NH2 -terminal kinase 1/ 2 signalling pathways, Clin. Exp. Immun, 162-172.
  • the JAK-3 inhibitor CP-690550 is a potent anti-inflammatory agent in a murine model of pulmonary eosinophilia, Eur J Pharmaco 154-161. Mullighan CG, Zhang J, Harvey RC, Collins-Underwood JR, Schulman BA, Phillips LA, Tasian SK, Loh ML,

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne de nouveaux composés [1,2,4]-triazolo-[1,5-a]-pyridine qui ont la formule représentée par la formule I. Les composés peuvent être préparés sous forme de composition pharmaceutique, et peuvent être utilisés pour la prévention et le traitement d'une variété d'états chez des mammifères y compris des humains, comprenant, en tant qu'exemple non limitant, des maladies comprenant la dégradation du cartilage, la dégradation osseuse et/ou articulaire, par exemple l'ostéoarthrite ; et/ou des états comprenant des réponses inflammatoires ou immunitaires, telles que la maladie de Crohn, l’arthrite rhumatoïde, le psoriasis, une maladie allergique des voies respiratoires (par exemple l'asthme, la rhinite), l’arthrite idiopathique juvénile, la rectocolite, les maladies inflammatoires de l'intestin, des états pathologiques induits par l'endotoxine (par exemple des complications après une chirurgie de pontage ou des états chroniques liés à l'endotoxine contribuant par exemple une insuffisance cardiaque chronique), des maladies comprenant une incapacité de renouvellement du cartilage (par exemple des maladies comprenant la stimulation anabolique de chondrocytes), des malformations congénitales du cartilage, des maladies associées à une hypersécrétion de l'IL6 et un rejet de la transplantation (par exemple un rejet d'un organe transplanté) et des maladies prolifératives.
PCT/EP2009/059600 2008-07-25 2009-07-24 Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires WO2010010188A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13593608P 2008-07-25 2008-07-25
US61/135,936 2008-07-25

Publications (1)

Publication Number Publication Date
WO2010010188A1 true WO2010010188A1 (fr) 2010-01-28

Family

ID=41056849

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/059600 WO2010010188A1 (fr) 2008-07-25 2009-07-24 Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires

Country Status (1)

Country Link
WO (1) WO2010010188A1 (fr)

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100093698A1 (en) * 2008-09-08 2010-04-15 Sogole Bahmanyar Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
WO2010092041A1 (fr) * 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines servant d'inhibiteurs de kinases
WO2011086099A1 (fr) 2010-01-15 2011-07-21 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Nouveaux dérivés de triazole substitués comme modulateurs de gamma secrétase
WO2011092272A1 (fr) * 2010-02-01 2011-08-04 F. Hoffmann-La Roche Ag Modulateurs de gamma-secrétase
WO2012025186A1 (fr) 2010-08-27 2012-03-01 Merck Patent Gmbh Dérivés triazolopyrazine
WO2012126984A1 (fr) 2011-03-24 2012-09-27 Janssen Pharmaceuticals, Inc. Nouveaux dérivés de triazolylpipérazine et triazolylpipéridine substitués à titre de modulateurs de gamma sécrétases
US8288403B2 (en) 2008-11-10 2012-10-16 Hoffmann-La Roche Inc. Heterocyclic gamma secretase modulators
WO2013010904A1 (fr) 2011-07-15 2013-01-24 Janssen Pharmaceuticals, Inc. Nouveaux dérivés d'indole substitués utilisés en tant que modulateurs de la gamma-sécrétase
AU2011211410B2 (en) * 2009-02-13 2013-01-31 Fovea Pharmaceuticals Sa [1,2,4] triazolo [1,5-A] pyridines as kinase inhibitors
US8389717B2 (en) 2008-10-09 2013-03-05 Hoffmann-La Roche Inc. Modulators for amyloid beta
US8486967B2 (en) 2010-02-17 2013-07-16 Hoffmann-La Roche Inc. Heteroaryl substituted piperidines
US8501936B2 (en) 2009-06-05 2013-08-06 Cephalon, Inc. Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US8546440B2 (en) 2008-12-18 2013-10-01 Janssen Pharmaceuticals, Inc. Substituted bicyclic imidazole derivatives as gamma secretase modulators
US8563545B2 (en) 2009-06-26 2013-10-22 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8772504B2 (en) 2009-02-19 2014-07-08 Janssen Pharmaceuticals, Inc. Substituted benzoxazole, benzimidazole, oxazolopyridine and imidazopyridine derivatives as gamma secretase modulators
US8796457B2 (en) 2009-06-26 2014-08-05 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8835482B2 (en) 2009-05-07 2014-09-16 Janssen Pharmaceuticals, Inc. Substituted indazole and aza-indazole derivatives as gamma secretase modulators
US8853240B2 (en) 2008-07-25 2014-10-07 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US8889673B2 (en) 2008-06-20 2014-11-18 Genentech, Inc. Triazolopyridine JAK inhibitor compounds and methods
US8946426B2 (en) 2009-02-06 2015-02-03 Janssen Pharmaceuticals, Inc. Substituted bicyclic heterocyclic compounds as gamma secretase modulators
US8946266B2 (en) 2009-07-15 2015-02-03 Janssen Pharmaceuticals, Inc. Substituted triazole and imidazole derivatives as gamma secretase modulators
US8962834B2 (en) 2008-02-22 2015-02-24 Hoffmann-La Roche Inc. Modulators of amyloid beta
US9181245B2 (en) 2012-05-16 2015-11-10 Janssen Pharmaceuticals, Inc. Substituted pyrido[1,2-a]pyrazines and substituted pyrido[1,2-a][1,4]diazepines for the treatment of (inter alia) Alzheimer's disease
US9434732B2 (en) 2008-06-20 2016-09-06 Genentech, Inc. Triazolopyridine JAK inhibitor compounds and methods
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
CN106188041A (zh) * 2016-06-29 2016-12-07 四川大学华西医院 [1,2,4]三唑并[1,5‑a]吡啶衍生物
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US9873709B2 (en) 2013-09-05 2018-01-23 Genentech, Inc. Triazolopyridine compounds, compositions and methods of use thereof
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US10112943B2 (en) 2012-12-20 2018-10-30 Janssen Pharmaceutica Nv Substituted imidazoles as gamma secretase modulators
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
US10246454B2 (en) 2013-01-17 2019-04-02 Janssen Pharmaceutica Nv Substituted 3,4-dihydro-2H-pyrido[1,2-a]pyrazine-1,6-diones as gamma secretase modulators
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions for the treatment of inflammatory disorders
US10562897B2 (en) 2014-01-16 2020-02-18 Janssen Pharmaceutica Nv Substituted 3,4-dihydro-2H-pyrido[1,2-a]pyrazine-1,6-diones as gamma secretase modulators
WO2020092015A1 (fr) 2018-11-02 2020-05-07 University Of Rochester Atténuation thérapeutique d'une infection épithéliale
US10708263B2 (en) 2014-02-07 2020-07-07 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
WO2021023207A1 (fr) * 2019-08-06 2021-02-11 江苏柯菲平医药股份有限公司 Inhibiteur de janus kinases jak et son utilisation
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
WO2021107590A1 (fr) * 2019-11-25 2021-06-03 주식회사 대웅제약 Nouveau dérivé de triazolopyridine et composition pharmaceutique le comprenant
CN115819425A (zh) * 2022-12-22 2023-03-21 五邑大学 一种三唑并六元氮杂环-2-胺类化合物及其制备方法和应用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003010167A1 (fr) * 2001-07-20 2003-02-06 F. Hoffmann-La Roche Ag Derives de 8-methoxy-(1,2,4)triazolo(1,5-a)pyridine et utilisations de ceux-ci sous la forme de ligands de recepteur d'adenosine
US20050222171A1 (en) * 2004-01-22 2005-10-06 Guido Bold Organic compounds
WO2007009773A1 (fr) * 2005-07-21 2007-01-25 Novartis Ag Derives de pyrazolo[1,5a]pyrimidin-7-yl-amine utilises en tant qu'inhibiteurs des proteines kinases
WO2008025821A1 (fr) * 2006-08-30 2008-03-06 Cellzome Limited Dérivés de triazole en tant qu'inhibiteurs de kinase
WO2009010530A1 (fr) * 2007-07-18 2009-01-22 Novartis Ag Composés hétéroaryliques bicycliques et leur utilisation comme inhibiteurs de kinase
WO2009017954A1 (fr) * 2007-08-01 2009-02-05 Phenomix Corporation Inhibiteurs de kinase jak2
WO2009027283A1 (fr) * 2007-08-31 2009-03-05 Merck Serono S.A. Composés de triazolopyridine et leur utilisation comme inhibiteurs de ask
WO2009047514A1 (fr) * 2007-10-10 2009-04-16 Cancer Research Technology Limited Composés [1,2,4]triazolo[1,5-a]pyridine et [1,2,4]triazolo[1,5-c]pyrimidine et leur utilisation

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003010167A1 (fr) * 2001-07-20 2003-02-06 F. Hoffmann-La Roche Ag Derives de 8-methoxy-(1,2,4)triazolo(1,5-a)pyridine et utilisations de ceux-ci sous la forme de ligands de recepteur d'adenosine
US20050222171A1 (en) * 2004-01-22 2005-10-06 Guido Bold Organic compounds
WO2007009773A1 (fr) * 2005-07-21 2007-01-25 Novartis Ag Derives de pyrazolo[1,5a]pyrimidin-7-yl-amine utilises en tant qu'inhibiteurs des proteines kinases
WO2008025821A1 (fr) * 2006-08-30 2008-03-06 Cellzome Limited Dérivés de triazole en tant qu'inhibiteurs de kinase
WO2009010530A1 (fr) * 2007-07-18 2009-01-22 Novartis Ag Composés hétéroaryliques bicycliques et leur utilisation comme inhibiteurs de kinase
WO2009017954A1 (fr) * 2007-08-01 2009-02-05 Phenomix Corporation Inhibiteurs de kinase jak2
WO2009027283A1 (fr) * 2007-08-31 2009-03-05 Merck Serono S.A. Composés de triazolopyridine et leur utilisation comme inhibiteurs de ask
WO2009047514A1 (fr) * 2007-10-10 2009-04-16 Cancer Research Technology Limited Composés [1,2,4]triazolo[1,5-a]pyridine et [1,2,4]triazolo[1,5-c]pyrimidine et leur utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NETTEKOVEN MATTHIAS ET AL: "Synthetic access to 2-amido-5-aryl-8-methoxy-triazolopyridine and 2-amido-5-morpholino-8-methoxy-triazolopyridine derivatives as potential inhibitors of the adenosine receptor subtypes", SYNTHESIS, GEORG THIEME VERLAG, STUTTGART, DE, no. 11, 1 January 2003 (2003-01-01), pages 1649 - 1652, XP002416826, ISSN: 0039-7881 *

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8962834B2 (en) 2008-02-22 2015-02-24 Hoffmann-La Roche Inc. Modulators of amyloid beta
US8889673B2 (en) 2008-06-20 2014-11-18 Genentech, Inc. Triazolopyridine JAK inhibitor compounds and methods
US9434732B2 (en) 2008-06-20 2016-09-06 Genentech, Inc. Triazolopyridine JAK inhibitor compounds and methods
US8853240B2 (en) 2008-07-25 2014-10-07 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US10206907B2 (en) 2008-07-25 2019-02-19 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US9415037B2 (en) 2008-07-25 2016-08-16 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US20100093698A1 (en) * 2008-09-08 2010-04-15 Sogole Bahmanyar Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
US8299056B2 (en) * 2008-09-08 2012-10-30 Signal Pharmaceuticals, Llc Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
US8389717B2 (en) 2008-10-09 2013-03-05 Hoffmann-La Roche Inc. Modulators for amyloid beta
US8288403B2 (en) 2008-11-10 2012-10-16 Hoffmann-La Roche Inc. Heterocyclic gamma secretase modulators
US8546440B2 (en) 2008-12-18 2013-10-01 Janssen Pharmaceuticals, Inc. Substituted bicyclic imidazole derivatives as gamma secretase modulators
US8946426B2 (en) 2009-02-06 2015-02-03 Janssen Pharmaceuticals, Inc. Substituted bicyclic heterocyclic compounds as gamma secretase modulators
WO2010092041A1 (fr) * 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines servant d'inhibiteurs de kinases
AU2011211410B2 (en) * 2009-02-13 2013-01-31 Fovea Pharmaceuticals Sa [1,2,4] triazolo [1,5-A] pyridines as kinase inhibitors
US8772504B2 (en) 2009-02-19 2014-07-08 Janssen Pharmaceuticals, Inc. Substituted benzoxazole, benzimidazole, oxazolopyridine and imidazopyridine derivatives as gamma secretase modulators
US8835482B2 (en) 2009-05-07 2014-09-16 Janssen Pharmaceuticals, Inc. Substituted indazole and aza-indazole derivatives as gamma secretase modulators
US8501936B2 (en) 2009-06-05 2013-08-06 Cephalon, Inc. Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US8633173B2 (en) 2009-06-05 2014-01-21 Cephalon, Inc Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US11000528B2 (en) 2009-06-26 2021-05-11 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8563545B2 (en) 2009-06-26 2013-10-22 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US10328081B2 (en) 2009-06-26 2019-06-25 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8796457B2 (en) 2009-06-26 2014-08-05 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US9505754B2 (en) 2009-06-26 2016-11-29 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8946266B2 (en) 2009-07-15 2015-02-03 Janssen Pharmaceuticals, Inc. Substituted triazole and imidazole derivatives as gamma secretase modulators
WO2011086098A1 (fr) 2010-01-15 2011-07-21 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Nouveaux dérivés de triazole bicycliques substitués comme modulateurs de gamma secrétase
WO2011086099A1 (fr) 2010-01-15 2011-07-21 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Nouveaux dérivés de triazole substitués comme modulateurs de gamma secrétase
US9079886B2 (en) 2010-01-15 2015-07-14 Janssen Pharmaceuticals, Inc. Substituted triazole derivatives as gamma secretase modulators
US9145399B2 (en) 2010-01-15 2015-09-29 Janssen Pharmaceuticals, Inc. Substituted bicyclic triazole derivatives as gamma secretase modulators
WO2011092272A1 (fr) * 2010-02-01 2011-08-04 F. Hoffmann-La Roche Ag Modulateurs de gamma-secrétase
JP2013518082A (ja) * 2010-02-01 2013-05-20 エフ.ホフマン−ラ ロシュ アーゲー γセクレターゼモジュレーター
US8486967B2 (en) 2010-02-17 2013-07-16 Hoffmann-La Roche Inc. Heteroaryl substituted piperidines
WO2012025186A1 (fr) 2010-08-27 2012-03-01 Merck Patent Gmbh Dérivés triazolopyrazine
WO2012126984A1 (fr) 2011-03-24 2012-09-27 Janssen Pharmaceuticals, Inc. Nouveaux dérivés de triazolylpipérazine et triazolylpipéridine substitués à titre de modulateurs de gamma sécrétases
US8987276B2 (en) 2011-03-24 2015-03-24 Janssen Pharmaceuticals, Inc. Substituted triazolyl piperazine and triazolyl piperidine derivatives as gamma secretase modulators
US9115143B2 (en) 2011-07-15 2015-08-25 Janssen Pharmaceuticals, Inc. Substituted indole derivatives as gamma secretase modulators
WO2013010904A1 (fr) 2011-07-15 2013-01-24 Janssen Pharmaceuticals, Inc. Nouveaux dérivés d'indole substitués utilisés en tant que modulateurs de la gamma-sécrétase
US9181245B2 (en) 2012-05-16 2015-11-10 Janssen Pharmaceuticals, Inc. Substituted pyrido[1,2-a]pyrazines and substituted pyrido[1,2-a][1,4]diazepines for the treatment of (inter alia) Alzheimer's disease
US10112943B2 (en) 2012-12-20 2018-10-30 Janssen Pharmaceutica Nv Substituted imidazoles as gamma secretase modulators
US10246454B2 (en) 2013-01-17 2019-04-02 Janssen Pharmaceutica Nv Substituted 3,4-dihydro-2H-pyrido[1,2-a]pyrazine-1,6-diones as gamma secretase modulators
US9873709B2 (en) 2013-09-05 2018-01-23 Genentech, Inc. Triazolopyridine compounds, compositions and methods of use thereof
US10562897B2 (en) 2014-01-16 2020-02-18 Janssen Pharmaceutica Nv Substituted 3,4-dihydro-2H-pyrido[1,2-a]pyrazine-1,6-diones as gamma secretase modulators
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions for the treatment of inflammatory disorders
US11667633B2 (en) 2014-02-07 2023-06-06 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US10708263B2 (en) 2014-02-07 2020-07-07 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US10919890B2 (en) 2014-02-07 2021-02-16 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US9527835B2 (en) 2014-02-13 2016-12-27 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10717737B2 (en) 2014-02-13 2020-07-21 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9994546B2 (en) 2014-02-13 2018-06-12 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11155532B2 (en) 2014-02-13 2021-10-26 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10676457B2 (en) 2014-02-13 2020-06-09 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11247992B2 (en) 2014-02-13 2022-02-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10513493B2 (en) 2014-02-13 2019-12-24 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10174030B2 (en) 2014-02-13 2019-01-08 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493442B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10300051B2 (en) 2014-02-13 2019-05-28 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9493450B2 (en) 2014-02-13 2016-11-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10125133B2 (en) 2014-07-10 2018-11-13 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyridines and substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10556908B2 (en) 2014-07-10 2020-02-11 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10138249B2 (en) 2014-07-10 2018-11-27 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10640503B2 (en) 2014-07-10 2020-05-05 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10968221B2 (en) 2014-07-10 2021-04-06 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10112950B2 (en) 2014-07-10 2018-10-30 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10047086B2 (en) 2014-07-10 2018-08-14 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US10800779B2 (en) 2015-04-03 2020-10-13 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US11401272B2 (en) 2015-04-03 2022-08-02 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US11498900B2 (en) 2015-08-12 2022-11-15 Incyte Corporation Salts of an LSD1 inhibitor
US10723700B2 (en) 2015-08-12 2020-07-28 Incyte Corporation Salts of an LSD1 inhibitor
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
CN106188041A (zh) * 2016-06-29 2016-12-07 四川大学华西医院 [1,2,4]三唑并[1,5‑a]吡啶衍生物
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11512064B2 (en) 2018-08-31 2022-11-29 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
WO2020092015A1 (fr) 2018-11-02 2020-05-07 University Of Rochester Atténuation thérapeutique d'une infection épithéliale
WO2021023207A1 (fr) * 2019-08-06 2021-02-11 江苏柯菲平医药股份有限公司 Inhibiteur de janus kinases jak et son utilisation
WO2021107590A1 (fr) * 2019-11-25 2021-06-03 주식회사 대웅제약 Nouveau dérivé de triazolopyridine et composition pharmaceutique le comprenant
CN114728963A (zh) * 2019-11-25 2022-07-08 株式会社大熊制药 新型三唑吡啶衍生物和包括其的药物组合物
JP2023503875A (ja) * 2019-11-25 2023-02-01 デウン ファーマシューティカル カンパニー リミテッド 新規なトリアゾロピリジン誘導体およびこれを含む薬学組成物
JP7371253B2 (ja) 2019-11-25 2023-10-30 デウン ファーマシューティカル カンパニー リミテッド 新規なトリアゾロピリジン誘導体およびこれを含む薬学組成物
CN114728963B (zh) * 2019-11-25 2023-10-31 株式会社大熊制药 新型三唑吡啶衍生物和包括其的药物组合物
CN115819425A (zh) * 2022-12-22 2023-03-21 五邑大学 一种三唑并六元氮杂环-2-胺类化合物及其制备方法和应用

Similar Documents

Publication Publication Date Title
AU2009273144B2 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010188A1 (fr) Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires
WO2010010189A1 (fr) Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires
CA2833963C (fr) Nouveau compose utile pour le traitement de maladies degeneratives et inflammatoires
WO2010010184A1 (fr) [1, 2, 4]triazolo[1, 5-a]pyridines utilisées comme inhibiteurs de jak
WO2010010187A1 (fr) Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires
WO2010010186A1 (fr) Nouveaux composés utiles pour le traitement de maladies dégénératives et inflammatoires
EP2445911A1 (fr) 5-phenyl-[1,2,4]triazolo[1,5-a]pyridin-2-yl-carboxamides utilises en tant qu'inhibiteurs de jak
CA2765988A1 (fr) Acide cyclopropanecarboxylique {5[-4-(3,3-dimethyl-azetidine-1-carbonyl)-phenyl]-[1,2,4]triazolo[1,5-a]pyridin-2-yl}-amide utile pour l'inhibition de janus kinase (jak)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09781069

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09781069

Country of ref document: EP

Kind code of ref document: A1