WO2010010190A1 - Novel compounds useful for the treatment of degenerative and inflammatory diseases - Google Patents

Novel compounds useful for the treatment of degenerative and inflammatory diseases Download PDF

Info

Publication number
WO2010010190A1
WO2010010190A1 PCT/EP2009/059604 EP2009059604W WO2010010190A1 WO 2010010190 A1 WO2010010190 A1 WO 2010010190A1 EP 2009059604 W EP2009059604 W EP 2009059604W WO 2010010190 A1 WO2010010190 A1 WO 2010010190A1
Authority
WO
WIPO (PCT)
Prior art keywords
unsubstituted
substituted
compound
alkyl
cpc
Prior art date
Application number
PCT/EP2009/059604
Other languages
French (fr)
Inventor
Christel Jeanne Marie Menet
Luc Juliaan Corina Van Rompaey
Stephen Robert Fletcher
Javier Blanc
Nolwenn Jouannigot
Alastair James Hodges
Koen Kurt Smits
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
Priority to KR1020117004383A priority Critical patent/KR101711911B1/en
Priority to RS20140345A priority patent/RS53406B/en
Application filed by Galapagos Nv filed Critical Galapagos Nv
Priority to DK09781072.5T priority patent/DK2361251T3/en
Priority to EP09781072.5A priority patent/EP2361251B1/en
Priority to BRPI0916659A priority patent/BRPI0916659B8/en
Priority to SI200930973T priority patent/SI2361251T1/en
Priority to MX2011000758A priority patent/MX2011000758A/en
Priority to US13/055,936 priority patent/US8853240B2/en
Priority to EA201170257A priority patent/EA018587B1/en
Priority to CN200980129088.4A priority patent/CN102105471B/en
Priority to AU2009273143A priority patent/AU2009273143B2/en
Priority to JP2011519188A priority patent/JP5559168B2/en
Priority to CA2730757A priority patent/CA2730757A1/en
Priority to MEP-2014-72A priority patent/ME02046B/en
Priority to ES09781072.5T priority patent/ES2485913T3/en
Priority to PL09781072T priority patent/PL2361251T3/en
Publication of WO2010010190A1 publication Critical patent/WO2010010190A1/en
Priority to IL210261A priority patent/IL210261A/en
Priority to ZA2011/00396A priority patent/ZA201100396B/en
Priority to HK11112990.2A priority patent/HK1158635A1/en
Priority to SM201400072T priority patent/SMT201400072B/en
Priority to HRP20140681AT priority patent/HRP20140681T1/en
Priority to US14/507,396 priority patent/US9415037B2/en
Priority to US15/236,578 priority patent/US10206907B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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

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/).
  • 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.
  • Rheumatoid arthritis is a chronic joint degenerative disease, characterized by inflammation and destruction of the joint structures. When the disease is unchecked, it leads to substantial disability and pain due to loss of joint functionality and even premature death. The aim of an RA therapy, therefore, is not to slow down the disease but to attain remission in order to stop the joint destruction. Besides the severity of the disease outcome, the high prevalence of RA ( ⁇ 0.8% of the adults are affected worldwide) means a high socio-economic impact. (For reviews on RA, we refer to Smolen and Steiner (2003); Lee and Weinblatt (2001); Choy and Panayi (2001); O'Dell (2004) and Firestein (2003)).
  • 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.
  • grade I osteoarthritis is reached; when the tangential surface area has disappeared, grade II osteoarthritis is reached.
  • degeneration and destruction which affect the deep and the calcified cartilage layers that border with the subchondral bone.
  • 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, 2006). Taken together, no disease modifying osteoarthritic drugs are available.
  • NSAIDs non-steroidal anti- inflammatory drugs
  • 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.
  • Other reported methods include application of tendinous, periosteal, fascial, muscular or perichondral grafts; implantation of fibrin or cultured chondrocytes; implantation of synthetic matrices, such as collagen, carbon fiber; administration of electromagnetic fields. All of these have reported minimal and incomplete effects, resulting in a poor quality tissue that can neither support the weighted load nor allow the restoration of an articular function with normal movement.
  • 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.
  • Insulin-like growth factor-I IGF-I is the predominant anabolic growth factor in synovial fluid and stimulates the synthesis of both proteoglycans and collagen.
  • 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 ef ⁇ /., 1998).
  • Oncostatin M induces MMP and TIMP3 gene expression in primary chondrocytes by activation of JAK/STAT and MAPK signaling pathways.
  • Osaki et al. (2003) showed that interferon- gamma mediated inhibition of collagen II in chondrocytes involves JAK-STAT signaling.
  • ILl -beta induces cartilage catabolism by reducing the expression of matrix components, and by inducing the expression of collagenases and inducible nitric oxide synthase (NOS2), which mediates the production of nitric oxide (NO).
  • 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.
  • diseases involving cartilage degradation, bone and/or joint degradation for example osteoarthritis
  • 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
  • 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.
  • diseases involving cartilage degradation, bone and/or joint degradation for example osteoarthritis
  • 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
  • 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.
  • CyI is selected from aryl and heteroaryl
  • R 3b is independently selected from substituted or unsubstituted aryl, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5-10 membered heteroaryl; or R 3b is independently selected from O-R 3c , NH- R 3c , CO-R 3c , and CON(R 4a )-R 3c ; and R 3c is independently selected from substituted Ci-C 6 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5- 10 membered heteroaryl; each R 4a , R 4b and R 4c is independently selected from H, Ci-C 6 alkyl, substituted Ci-C 6 alkyl, C
  • l,2,4-triazolo[l,5-a]pyridine compounds according to Formula I are disclosed that are capable of capable of modulating the activity of JAK in vivo.
  • 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.
  • 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 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 described herein.
  • 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 compounds according to Formula I or a pharmaceutically acceptable salt thereof in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases.
  • diseases involving cartilage degradation, bone and/or joint degradation for example osteoarthritis
  • conditions involving inflammation or immune responses
  • 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.
  • diseases associated with JAK activity such as diseases involving cartilage degradation, bone and/or joint degradation, for example osteoarthritis
  • inflammation or immune responses such as Crohn's disease, rheumatoid arthritis, psoriasis, allergic airways disease (e.g. asthma, rhinitis), juvenile idiopathic arthritis, colitis
  • 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. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure
  • organ transplant rejection e.g. complications after bypass surgery or chronic endotoxin states contributing to e.g. chronic cardiac failure
  • cartilage, bone and/or joint degradation or degeneration such as osteoarthritis or cancers (e.g. solid tumours or leukaemias).
  • analogue means one analogue or more than one analogue.
  • 'Acyl' refers to a radical -C(O)R 20 , where R 20 is hydrogen, C 1 -C 8 alkyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 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-
  • Substituted Acyl' refers to a radical -C(O)R 21 , wherein R 21 is independently • CpC 8 alkyl, substituted with halo or hydroxy; or
  • 'Acylamino' refers to a radical -NR 22 C(O)R 23 , where R 22 is hydrogen, CpC 8 alkyl, C 3 -
  • Exemplary 'acylamino' include, but are not limited to, formylamino, acetylamino, cyclohexylcarbonylamino, cyclohexylmethyl-carbonylamino, benzoylamino and benzylcarbonylamino.
  • Exemplary 'acylamino' groups are -NR 21 C(O)-Ci-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 -Ci 0 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 CpC 8 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
  • 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 ⁇ -Cio 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 -0-(CH 2 )t(C 6 -Cio aryl), -O-(CH 2 ) t (5-10 membered heteroaryl), -0-(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 CpC 4 haloalkyl, unsubstituted CpC 4 hydroxyalkyl, or unsubstituted CpC 4 haloalkoxy or hydroxy.
  • Particular exemplary 'substituted alkoxy' groups are OCF 3 , OCH 2 CF 3 , OCH 2 Ph, OCH 2 - cyclopropyl, OCH 2 CH 2 OH, OCH 2 CH 2 NMe 2 .
  • Alkoxycarbonyl' refers to a radical -C(O)-OR 27 where R 27 represents an Ci-C 8 alkyl
  • 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 (-0-C(O)-NR 2) , aminosulfonyl,
  • 'substituted alkyl' refers to a CpCg 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, Ci-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 heterocycloal
  • '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 ) 2 where each R is independently selected from:
  • -N(R ) 2 is an amino group.
  • exemplary 'substituted amino' groups are -NR 33' -CpC 8 alkyl, -NR 33' -(CH 2 ) t (C 6 -Ci 0 aryl), -NR 33' -(CH 2 ) t (5-10 membered heteroaryl), -NR 33' - (CH 2 MC 3 -Ci 0 cycloalkyl), and -NR -(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 unsubstituted CpC 4 alkyl,
  • Alkylamino' refers to the group -NHR 34 , wherein R 34 is CpC 8 alkyl.
  • Substituted Alkylamino' refers to the group -NHR 35 , wherein R 35 is CpC 8 alkyl; and the alkyl group is substituted with halo, substituted or unsubstituted amino, hydroxy, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, 5-10 membered heteroaryl, aralkyl or heteroaralkyl; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted CpC 4 haloalkyl, unsubstituted
  • 'Dialkylamino' refers to the group -NR 42 R 43 , wherein each of R 42 and R 43 are independently selected from CpCg alkyl.
  • Dialkylamino' refers to the group -NR 44 R 45 , wherein each of R 44 and R 45 are independently selected from CpCg alkyl; and the alkyl group is independently substituted with halo, hydroxy, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, 5-10 membered heteroaryl, aralkyl or heteroaralkyl; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted
  • Cp 4 haloalkyl unsubstituted CpC 4 hydroxyalkyl, or unsubstituted CpC 4 haloalkoxy or hydroxy.
  • '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:
  • '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 10. Where the 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, triphenylene and trinaphthalene.
  • aryl groups include phenyl, naphthyl, indenyl, and tetrahydronaphthyl.
  • '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, CpC 8 alkyl, CpC 8 haloalkyl, CpC 8 haloalkoxy, cyano, hydroxy, CpC 8 alkoxy, and amino.
  • Examples of representative substituted aryls include the following
  • R 49 and R 50 may be hydrogen and at least one of R 49 and R 50 is each independently selected from CpC 8 alkyl, 4-10 membered heterocycloalkyl, CpC 8 alkoxy, hetero- O-aryl, alkylamino, 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, S0 2 alkyl, Saryl, SOaryl, S0 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 (saturated or unsaturated) from 5 to 8 atoms, optionally
  • R 51 , and R 52 are independently hydrogen, CpC 8 alkyl, CpC 4 haloalkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 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 CpC 4 alkyl, halo, cyano, unsubstituted CpC 4 alkoxy, unsubstituted Cp 4 haloalkyl, unsubstituted CpC 4 hydroxyalkyl, or unsubstituted CpC 4 haloalkoxy or hydroxy.
  • 'Azido' refers to the radical -N 3 .
  • 'Amido' refers to the radical -C(O)NH 2 .
  • Exemplary 'Substituted Amido ' groups are -C(O) NR 53' -C r C 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(0)NR 53' -(CH 2 )t(C 3 -Cio 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 CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted CpC 4 hal
  • '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.
  • Particular 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:
  • 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 54 is independently hydrogen, CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, and 5-10 membered heteroaryl.
  • R 54 is independently hydrogen, CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, and 5-10 membered heteroaryl.
  • Examples of representative heteroaryls include the following:
  • each Y is selected from carbonyl, N, NR 55 , O and S; and R 55 is independently hydrogen, CpC 8 alkyl, C3-C 1 0 cycloalkyl, 4-10 membered heterocycloalkyl, C ⁇ -Cio 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.
  • 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, CpC 8 alkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C ⁇ -Cio 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 2 ), aminocarbonylamino (-NR -C(O)- NR 2
  • 'Nitro' refers to the radical -NO 2 .
  • 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(O)NR R “ , -NR “ R “ , -(CR '” R '” ) m OR '” , wherein, each R " is independently selected from H, CpC 8 alkyl, -(CH 2 ) t (C 6 -Ci 0 aryl), -(CH 2 ) t (5-10 membered heteroaryl), -(CH 2 ) t (C 3 -Ci 0 cycloalkyl), and -(CH 2 ) t (4-10 membered heterocycloalky
  • any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present may themselves be substituted by unsubstituted CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted CpC 4 haloalkyl, unsubstituted CpC 4 hydroxyalkyl, or unsubstituted CpC 4 haloalkoxy or hydroxy.
  • Each R independently represents H or CpC 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-(CH 2 )t(C 6 -Cio aryl), -S-(CH 2 ) t (5-10 membered heteroaryl), -S-(CH 2 ) t (C 3 -Ci 0 cycloalkyl), and -S-(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
  • Substituted sulfanyl' includes the groups 'alkylsulfanyl' or 'alkylthio', 'substituted alkylthio' or 'substituted alkylsulfanyl', 'cycloalkylsulfanyl' or 'cycloalkylthio', 'substituted cycloalkylsulfanyl' or 'substituted cycloalkylthio', 'arylsulfanyl' or 'arylthio' and 'heteroarylsulfanyl' or 'heteroarylthio' as defined below.
  • Substituted sulfinyl' refers to the group -S(O)R 68 , wherein R 68 is selected from:
  • 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 CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted CpC 4 haloalkyl, unsubstit
  • substituted sulfinyl includes the groups 'alkylsulfinyl', 'substituted alkylsulfinyP, 'cycloalkylsulfinyl', 'substituted cycloalkylsulfinyP, 'arylsulfinyP and 'heteroarylsulfmyP as defined herein.
  • 'Substituted sulfonyP refers to the group -S(O) 2 R 75 , wherein R 75 is selected from:
  • Exemplary 'substituted sulfonyP groups are -S(O) 2 -(CpC 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 - Ci 0 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 CpC 4 alkyl, halo, unsubstituted CpC 4 alkoxy, unsubstituted CpC
  • substituted sulfonyl includes the groups alkylsulfonyl, substituted alkylsulfonyl, cycloalkylsulfonyl, substituted cycloalkylsulfonyl, arylsulfonyl and heteroarylsulfonyl.
  • 'Sulfo' or 'sulfonic acid' refers to a radical such as -SO3H.
  • '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 -S(O) 2 -O-(C 3 -Ci 0 cycloalkyl), -S(O) 2 -O-(CH 2 MC 6 -C 10 aryl), -S(O) 2 -O-(CH 2 ) t (5-10 membered heteroaryl), -S(O) 2 -O-(CH 2 X(C 3 -Ci 0 cycloalkyl), and -S(O) 2 -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 CpC 4 alkyl, hal
  • heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
  • 'Pharmaceutically acceptable means approved or approvable by a regulatory agency of the Federal or a state government or the corresponding agency in countries other than the United States, or that is listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly, in humans.
  • '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-toluenesulfonic acid
  • 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. Conventional 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.
  • '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.
  • 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 anti-malarial 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.
  • '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 dermatitises, inflammatory bowel disease (e.g.
  • Atherosclerosis Crohn's disease and ulcerative colitis
  • amyotrophic lateral sclerosis Particularly the term refers to COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and 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.
  • pancreatic islets 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.
  • 'proliferative disease(s)' refers to conditions such as cancer
  • myeloproliferative disorders named in particular Re JAK2 activating mutations (polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis), leukemia (e.g. acute myeloid leukaemia and acute lymphoblastic leukemia), multiple myeloma, psoriasis, restenosis, sclerodermitis or fibrosis.
  • leukemia e.g. acute myeloid leukaemia and acute lymphoblastic leukemia
  • multiple myeloma psoriasis
  • restenosis sclerodermitis or fibrosis.
  • sclerodermitis or fibrosis.
  • cancer e.g. uterine leiomyosarcoma or prostate cancer
  • leukemia e.g. acute myeloid leukaemia and acute lymphoblastic leukemia
  • multiple myeloma psorias
  • 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.
  • metastatic rumour cell types such as but not limited to, melanoma, lymphoma, leukaemia, fibrosarcoma, rhabdomyosarcoma, and mastocytoma
  • 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, glioblast
  • 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).
  • Diseases involving the anabolic stimulation of chondrocytes includes conditions such as 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.
  • ranges are referred to herein, for example but without limitation, CpCg alkyl
  • the citation of a range should be considered a representation of each member of said range.
  • Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but in the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985).
  • 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 ( 2 H or D), carbon- 13 ( 13 C), nitrogen- 15 ( 15 N), or the like.
  • the following atoms, where present, may vary, so that for example, any hydrogen may be 2 H/D, any carbon may be 13 C, or any nitrogen may be 15 N, and that the presence and placement of such atoms may be determined within the skill of the art.
  • 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. 3 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
  • Stereoisomers that are not mirror images of one another are termed 'diastereomers' and those that are non-superimposable mirror images of each other are termed 'enantiomers'.
  • a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible.
  • 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 phenylnitromethane, 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.
  • 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.
  • diseases involving cartilage degradation, bone and/or joint degradation for example osteoarthritis
  • 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
  • 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 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.
  • CyI is selected from aryl and heteroaryl
  • R 3b is independently selected from substituted or unsubstituted aryl, substituted or unsubstituted C 3 -C 7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5-10 membered heteroaryl; or R 3b is independently selected from O-R 3c , NH- R 3c , CO-R 3c , and CON(R 4a )-R 3c ; and R 3c is independently selected from substituted C r C 6 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5- 10 membered heteroaryl; each R 4a , R 4b and R 4c is independently selected from H, Ci-C 6 alkyl, substituted Ci-C 6 alkyl,
  • the present invention provides compounds according to Formula I or a pharmaceutically acceptable salt thereof in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases.
  • diseases involving cartilage degradation, bone and/or joint degradation for example osteoarthritis
  • conditions involving inflammation or immune responses
  • CyI is selected from aryl and heteroaryl;
  • each R 1 is independently selected from unsubstituted CpC 6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted Ci-C 6 alkoxy, unsubstituted amido, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from unsubstituted Ci-Ce alkyl, unsubstituted acyl, unsubstituted acylamino, unsub
  • R a is selected from unsubstituted CpC 6 alkyl and unsubstituted C3-C7 cycloalkyl;
  • R 3b is independently selected from aryl (which aryl may be substituted with halo, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted C 1 -C 4 alkyl, unsubstituted C 1 -C 4 alkoxy, unsubstituted CpC 4 haloalkyl, unsubstituted CpC 4 haloalkoxy, unsubstituted 5-7- membered heteroaryl, unsubstituted acylamino, unsubstituted amino, cyano, -(CH 2 )i_ 4 - CN), C 3 -C 7 cycloalkyl (which C 3 -C 7 cycloalkyl may be substituted with cyano), 4-7 membered heterocycloalkyl (which 4-7 membered heterocycloalkyl may be substituted with CpC 4 alkyl (which CpC 4 alkyl may be substituted with aryl, heteroary
  • Ci-C ⁇ alkyl (which CpC 4 alkyl is substituted with aryl (which aryl may be substituted with halo, CN, unsubstituted CpC 4 alkyl, unsubstituted 5-7-membered heterocycloalkyl, unsubstituted 5-10 membered heteroaryl), 5-10 membered heteroaryl (which heteroaryl may be substituted with unsubstituted CpC 4 alkyl)), aryl (which aryl may be substituted with halo, CN, unsubstituted C 1 -C 4 alkoxy, unsubstituted amido, unsubstituted CpC 4 alkyl, unsubstituted CpC 4 , haloalkyl, -(CH 2 ) i_ 4 -CN), C 3 -C 7 cycloalkyl (which C 3 -C 7 cycloalkyl may be substituted with ), unsubstituted 4
  • CyI is selected from aryl and heteroaryl;
  • each R 1 is independently selected from unsubstituted CpC 6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted Ci-Ce alkoxy, unsubstituted amido, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from unsubstituted Ci-C ⁇ alkyl, unsubstituted acyl, unsubstituted acylamino, unsub
  • R a is selected from unsubstituted Ci-C ⁇ alkyl and unsubstituted C3-C7 cycloalkyl;
  • R 3b is independently selected from aryl (which aryl may be substituted with halo, unsubstituted
  • ml is 1 or 2; each R 1 is independently selected from Ci-C ⁇ alkyl, substituted Ci-C ⁇ alkyl, and halo.
  • ml is 1 or 2 and each R 1 is independently selected from Me, CF 3 , Cl and F.
  • R a is substituted or unsubstituted Ci-Ce alkyl.
  • R a is substituted or unsubstituted C3-C7 cycloalkyl.
  • R a is cyclopropyl, cyclobutyl, or cyclop entyl.
  • R 4b and R 4c are independently selected from H and Me.
  • CyI is Ph; and m2 is 0.
  • CyI is Ph; m2 is 1, 2 or
  • each R a is independently Ci-C ⁇ alkyl, Ci-Ce haloalkyl, Ci-C ⁇ alkoxy, or halo.
  • each R 3a is independently Cl, F, Me, Et, OMe, CF 3 , CONH 2 , CONMe 2 , CONHMe, CN,
  • CyI is 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 benzodioxanyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted quinolinyl, or substituted or unsubstituted isoquinolinyl; and
  • Ll, R 3b , and nl are as described for Formula I.
  • R 3b is substituted or unsubstituted aryl, substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted
  • R 3b is substituted or unsubstituted cyclopropyl, substituted or unsubstituted cyclobutyl, substituted or unsubstituted cyclohexyl, or substituted or unsubstituted cyclopentyl.
  • R 3b is substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrrolyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted tetrazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted oxadiazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted thiophenyl, substituted or unsubstituted indolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted benzimidazoly
  • N(R 4a )CO-; nl is 0, 1, 2, 3, or 4; and R 3b is substituted or unsubstituted 4-7-membered heterocycloalkyl, provided that when the heterocycle is attached via a heteroatom, and Ll is -O-, -N(R 4a )-, -SO 2 N(R 4a )-, and -CON(R 4a )-, nl is not 0 or 1.
  • Ll is a single bond.
  • Ll is selected from -O-, and -N(R 4a )-.
  • Ll is selected from -C(O)-, and -S(O) 2 -.
  • Ll is selected from -CON(R 4a )-, and -SO 2 N(R 4a )-.
  • Ll is selected from -N(R 4a )SO 2 - and -N(R 4a )CO-.
  • Ll is -
  • R 4a is H, substituted or unsubstituted CpC 4 alkyl, substituted or unsubstituted CpC 4 alkoxy.
  • R 4a is H
  • R 4a is -(CH 2 ) n2 -R 6a ; wherein n 2 is 0, 1, 2 and R 6a is H, CN, NMe 2 , or tetrahydrofuranyl.
  • R 4a is -CH(CH 3 )-
  • R 3b is OPh,and O-(4-
  • R 3b is CO-R 3c ; and R 3c is:
  • the compound is according to formula III, and -Ph-Ll -(CH 2 ) n i-R 3 is:
  • n2 is nl; and R 3b , and nl are as described for Formula 1 ; and Cy3 is a substituted or unsubstituted nitrogen containing 4-7-membered heterocycloalkyl group.
  • the compound is according to Formula III, and -Ph-Ll -(CH 2 ) n i-
  • R 3b is:
  • n2 is nl; and R , 3b , and nl are as described for Formula 1; and Cy3 is a substituted or unsubstituted nitrogen containing 4-7-membered heterocycloalkyl group.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and R 3b is unsubstituted aryl.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and R 3b is substituted aryl.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and R 3b is
  • 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, CpCg alkoxy, hetero-O-aryl, alkylamino,
  • 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, S0 2 alkyl, Saryl, SOaryl, S0 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 51 , and R 52 are independently hydrogen, Ci-Cg alkyl, CpC 4 haloalkyl, C 3 -Ci 0 cycloalkyl, 4-10 membered heterocycloalkyl, C 6 -Ci 0 aryl, substituted aryl, 5-10 membered heteroaryl.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and R 3b is aryl, substituted with one or more of groups selected from halo, CpCg alkyl, Cp
  • Ci-Cg haloalkoxy C 8 haloalkyl, Ci-Cg haloalkoxy, cyano, hydroxy, Ci-Cg alkoxy, and amino.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and R 3b is phenyl, substituted with one or more of groups selected from halo, Ci-Cg alkyl,
  • the-Ph-Ll-(CH 2 ) n pR 3b and R 3 b are as described in the preceding paragraph, and the substitution is other than 3-OMe.
  • the compound is other than N-[5-[4-[(3-methoxyphenyl)- methoxy]phenyl][l,2,4]triazolo[l,5-a]pyridin-2-yl]-cyclopropanecarboxamide.
  • the -Ph-Ll -(CH 2 ) n pR 3b group is as described in the preceding paragraphs, and R 3b is substituted or unsubstituted heteroaryl.
  • the -Ph-Ll -(CH 2 ) n pR 3b is as described in the preceding paragraphs, and R 3b is unsubstituted thiophenyl, pyrrolyl, benzothiophenyl, benzofuranyl, indolyl, pyridyl, quinolinyl, imidazolyl, oxazolyl and pyrazinyl
  • the -Ph-Ll -(CH 2 ) n pR 3b group is as described in the preceding paragraphs, and R 3b is thiophenyl, pyrrolyl, benzothiophenyl, benzofuranyl, indolyl, pyridyl, quinolinyl, imidazolyl, oxazoleyl and pyrazineyl, substituted with one or more groups selected from halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR "' SO 2 R " , -SO 2 NR R “ , -C(O)R “ , -C(O)OR " , -
  • each R " is independently selected from H, C 1 -C 8 alkyl, -(CH 2 MC 6 -C 10 aryl), -(CH 2 ) t (5-10 membered heteroaryl), -(CH 2 X(C 3 -Ci 0 cycloalkyl), and -(CH 2 ) t (4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4.
  • the -Ph-Ll -(CH 2 ) nr R 3b group is as described in the preceding paragraphs, and R 3b is unsubstituted pyridyl.
  • the -Ph-Ll -(CH 2 ) nr R 3b group is as described in the preceding paragraphs, and R 3b is pyridyl, substituted with one or more groups 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 “ , -
  • Ci-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
  • t is an integer from 0 to 4.
  • the -Ph-Ll -(CH 2 ) nr R 3b is as described in the preceding paragraphs, and R 3b is pyridyl, substituted with halo, cyano, methyl, or trifluoromethyl.
  • the -Ph-Ll -(CH 2 ) n i-R 3b is as described in the preceding paragraphs, and nl or n2 is 1.
  • the -Ph-Ll -(CH 2 ) nr R 3b is as described in the preceding paragraphs, and nl or n2 is 2.
  • the compound is according to Formula III and R 3b is selected from:
  • each R 5a is independently C 1 -C 4 alkyl, halo, CF 3 or Phenyl;
  • R 5b is H, aryl, 5-10 membered heteroaryl, heteroaryl, C 3 -C 6 cycloalkyl, or 4-7 membered heterocycloalkyl; and
  • m5 is 0, 1 or 2.
  • the compound is according to Formula III and R 3b is selected from: wherein each R 5a is independently C 1 -C 4 alkyl, halo, oxo, CF 3 or Phenyl; R 5b is H, C 1 -C 4 alkyl, aryl, 5-10 membered heteroaryl, heteroaryl, C3-C6 cycloalkyl, or 4-7 membered heterocycloalkyl; and m5 is 0, 1 or
  • the compound is according to Formula III and Cy3 is selected from:
  • each R 5a is independently C 1 -C 4 alkyl, halo, CF 3 or Phenyl;
  • R 5b is H, aryl, 5-10 membered heteroaryl heteroaryl, C 3 -C 6 cycloalkyl, or 4-7 membered heterocycloalkyl;
  • R 5c is H, or CpC 4 alkyl;
  • m5 is O, 1, or 2;
  • n5 is O, I, or 2.
  • the compound is according to any one of Formulae
  • IVa, IVb, IVc, IVd, IVe, or IVf
  • the compound is according to any one of Formulae
  • the compound is according to any one of Formulae
  • the compound is according to any one of Formulae
  • the compound is according to any one of
  • the compound is according to any one of
  • the present invention provides compounds according to Formula X, or XI:
  • R 3b is substituted or unsubstituted 4-7 membered heterocycloalkyl; provided that when the compound is according to Formula X, the heterocycloalkyl ring is other than unsubstituted morpholin-1- yi.
  • the compound is according to Formula X. In another embodiment, the compound is according to Formula XL
  • R 3b is unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, azepinyl, pyrrolidonyl, pyranyl, dihydrothiophenyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothiophenyl, dioxanyl, tetrahydropyranyl, imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 2-pyrazolineyl, pyrazolidinyl, thiomorpholinyl-S-oxide, and thiomorpholinyl-S,S-dioxide piperidonyl, or piperazonyl.
  • R 3b is unsubstituted azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, thiomorpholin-l-yl-S,S-dioxide, piperazin-1- yl, or azepin-1-yl.
  • R 3b is unsubstituted azetidin-1-yl.
  • R 3b is unsubstituted pyrrolidin-1-yl.
  • R 3b is unsubstituted piperidin-1-yl or piperazin-1-yl.
  • R 3b is unsubstituted thiomorpholin-1 -yl-S,S-dioxide.
  • R 3b is unsubstituted azepin-1-yl.
  • R 3b is azetidin-
  • R 3b is azetidin-
  • R 3b is unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, azepinyl, pyrrolidonyl, pyranyl, dihydrothiophenyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothiophenyl, dioxanyl, tetrahydropyranyl, imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 2-pyrazolineyl, pyrazolidinyl, morpholinyl, thiomorpholinyl-S-oxide, and thiomorpholinyl-
  • R 3b is unsubstituted azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholinyl, thiomorpholin-l-yl-S,S- dioxide, piperazin-1-yl, or azepin-1-yl.
  • R 3b is unsubstituted azetidin-1-yl.
  • R 3b is unsubstituted pyrrolidin-1-yl.
  • R 3b is unsubstituted piperidin-1-yl or piperazin-1-yl.
  • R 3b is unsubstituted thiomorpholin-1 -yl-S,S-dioxide.
  • R 3b is unsubstituted morpholin-1-yl.
  • R 3b is unsubstituted azepin-1-yl.
  • R 3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl, piperazin-1-yl, or azepin-1-yl; substituted with one or more group selected from C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, cyano, amino, dialkylamino, dialkylaminomethyl, hydroxy, halo, acyl, acylamino, C 1 -C 4 hydroxyalkyl, C 1 -C 4 alkoxy, carboxamido, and C 1 -C 4 dialkyl carboxamido.
  • R 3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl, piperazin-1-yl, or azepin-1-yl; substituted with Me, CF 3 , F, Cl, difluoro, dimethyl, hydroxy, cyano, dimethylamino, dimethylaminomethyl, hydroxymethyl, carboxamido, N,N-dimethylcarboxamido, methoxy, ethoxy, or 2,2,2,-trifluoroethyl.
  • the compound is selected from the compounds exemplified in Table 1.
  • the compound of the invention is not an isotopic variant.
  • 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 of the compound.
  • a compound of the invention according to any one of the embodiments herein described is a solvate of a pharmaceutically acceptable salt of the compound.
  • a compound of the invention includes one in which several or each embodiment in the above Formula, as well as other formulae presented herein, is selected from one or more of particular members or groups designated respectively, for each variable. Therefore, this invention is intended to include all combinations of such embodiments within its scope.
  • the present invention additionally 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.
  • 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 the 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 the invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal.
  • 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
  • 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 pref ⁇ lled, 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.
  • 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) may be added and the resulting mixture is stirred until it congeals.
  • the present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of JAK.
  • the 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 a 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.
  • osteoarthritis a condition associated with, or diseases involving the anabolic stimulation of chondrocytes
  • 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 therapeutically effective amount of a compound of the invention, or 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, 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 an effective amount of one or more of the compounds of the invention or 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 an effective amount of one or more of the compounds of the invention or 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. complications after bypass surgery or chronic endotoxin states contributing to e.g.
  • 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 methods comprise administering an effective amount of one or more of the compounds of the invention or 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 methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.
  • 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.
  • 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 methods comprise administering an effective amount of one or more of the compounds of the invention or 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 methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.
  • the invention provides methods of treating organ transplant rejection.
  • 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.
  • 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 particular 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 particular 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.
  • the regimen for treatment usually stretches over many months or years so oral dosing is preferred for patient convenience and tolerance. With oral dosing, one to five and especially two to four and typically three oral doses per day are representative regimens.
  • 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.
  • the compounds of this invention When used to prevent the onset of an inflammatory condition, the compounds of this 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 be safe and efficacious for such combined administration.
  • 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; particular agents include, but are not limited to, immunoregulatory agents e.g. azathioprine, corticosteroids (e.g.
  • prednisolone or dexamethasone cyclophosphamide
  • cyclosporin A tacrolimus
  • Mycophenolate Mofetil Mycophenolate Mofetil
  • muromonab-CD3 OKT3, e.g. Orthocolone®
  • ATG aspirin
  • acetaminophen ibuprofen
  • naproxen piroxicam
  • 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 cyclosporin), 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,
  • 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
  • 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. anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG)).
  • calcineurin inhibitors e.g. cyclosporin or tacrolimus (FK506)
  • mTOR inhibitors e.g. sirolimus,
  • 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.
  • 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. 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.
  • nonspecific beta-agonists injected or inhaled
  • injected or inhaled e.g. epinephrine, isoetharine, isoprotereno
  • 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 cyclosporin) 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 cyclosporin
  • 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. methot
  • 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,
  • a compound of the invention may be administered in combination with other therapies including, but not limited to phototherapy, or photochemotherapy (e.g. psoralen and ultraviolet A phototherapy (PUVA)).
  • photochemotherapy e.g. psoralen and ultraviolet A phototherapy (PUVA)
  • PUVA ultraviolet A phototherapy
  • 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 Cyl-Ll-(CR 4b R 4c ) nl -R 3b ; and CyI, Ll, nl, R 2a , R 3b , R 4b , and R 4c are as described herein.
  • R 4a , R 4b , R 4c , R 3b and nl are as described herein.
  • R 4a ,R 4b , R 4c ,R 3b and nl are as described herein.
  • Preparative HPLC Waters XBridge Prep Cl 8 5 ⁇ m ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H 2 O gradients. H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • R 4a ,R 4b , R 4c ,R 3b and nl are as described herein.
  • H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • Ar is Cyl-Ll-(CR 4b R 4c ) nl -R 3b ; and CyI, Ll, nl, R 3b , R 4b , and R 4c are as described herein.
  • H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • H 2 O contains either 0.1% TFA or 0.1% NH 3 .
  • R 4b , R 4c and R 3b and nl are as described herein.
  • the title compound is optained by method A using the intermediate boronate ester described above.
  • This compound was prepared via Method A using 3-(4-morpholinomethyl)- phenylboronic acid pinacol ester hydrochloride.
  • This compound was prepared via Method A using 2-(pyrrolidin-l-yl)pyrimidine-5- boronic acid pinacol ester.
  • This compound was prepared via Method A using 2-(4-morpholino)pyrimidine-5- boronic acid pinacol ester.
  • This compound was prepared via Method A using 2-(piperidin-l-yl)pyrimidme-5- boronic acid pinacol ester.
  • This compound was prepared via Method A using 4-(N- cyclopropylsulfonamide)phenylboronic acid pinacol ester.
  • This compound was prepared via Method A using piperidin-l-yl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-methanone.
  • This compound was prepared via Method A using pyrrolidin-l-yl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-methanone.
  • This compound was prepared via Method D using phenyl-acetyl chloride.
  • This compound was prepared via Method B using l-methyl-piperidin-4-ylamine.
  • This compound was prepared via Method D using pyrazine-2-carbonyl chloride.
  • This compound was prepared via Method C using iodocyclopentane.
  • This compound was prepared via Method D using cyclohexanecarbonyl chloride.
  • This compound was prepared via Method G using phenyl boronic acid.
  • This compound was prepared via Method C using 5-(chloromethyl)-l,3-dimethyl-lH- pyrazole.
  • This compound was prepared via Method D using 2-fluoro-phenyl sulfonyl chloride.
  • This compound was prepared via Method D using Pyridine-2-carboxylic acid (acid chloride formed by reaction of oxalyl chloride).
  • This compound was prepared via Method D using cyclopropanesulfonyl chloride.
  • This compound was prepared via Method C using 3-(chloromethyl)-l,5-dimethyl-lH- pyrazole.
  • This compound was prepared via Method C using 4-(chloromethyl)-2,5-dimethyl-l,3- oxazole.
  • This compound was prepared via Method D using pyridine-3-sulfonyl chloride.
  • This compound was prepared via Method D using l,3-dimethyl-lH-pyrazole-4-sulfonyl chloride.
  • This compound was prepared via Method D using 2-methyl-5-propyl-2H-pyrazole-3- carboxylic acid which was reacted first in presence of oxalyl chloride to obtain the acid chloride.
  • This compound was prepared via Method B using phenyl-piperidin-4-yl-methanone.
  • This compound was prepared via Method B using l-benzyl-[l,4]diazepane.
  • This compound was prepared via Method D using phenylsulfonyl chloride.
  • This compound was prepared via Method B using phenethylamine.
  • This compound was prepared via Method B using butyl-(3-morpholin-4-yl-propyl)- amine.
  • This compound was prepared via Method B using 4-(2-piperidin-4-yl-ethyl)- morpholine.
  • This compound was prepared via Method H using 4-(2-piperidin-4-yl-ethyl)- morpholine.
  • This compound was prepared via Method B using methyl-phenethyl-amine.
  • This compound was prepared via Method B using 2-(4-trifluoromethyl-phenyl)- ethylamine.
  • This compound was prepared via Method C using l-(2-bromo-ethyl)-lH-pyrazole.
  • This compound was prepared via Method B using 1 , 1 -dimethyl ⁇ -morpholin ⁇ -yl- ethylamine.
  • This compound was prepared via Method B using benzyl-methyl-piperidin-4-yl-amine.
  • This compound was prepared via Method B using [2-(3,4-dimethoxy-phenyl)-ethyl]- methyl-amine.
  • This compound was prepared via Method B using 2-(l-phenyl-lH-pyrazol-4-yl)- ethylamine.
  • This compound was prepared via Method A using 1 -benzyl- lH-pyrazole-4-boronic acid.
  • This compound was prepared via Method B using phenethylamine.
  • This compound was prepared via Method B using l-(3-chloro-phenyl)-piperazine.
  • This compound was prepared via Method B using 3 -phenyl-propylamine.
  • This compound was prepared via Method C using l-(4-benzyl-piperidin-l-yl)-2-chloro- ethanone.
  • This compound was prepared via Method C using 2-chloro-N-methyl-N-phenyl- acetamide.
  • This compound was prepared via Method H using phenethylamine.
  • This compound was prepared via Method C using the mesylate derivative of (4- pyrazol- 1 -yl-phenyl)-methanol.
  • This compound was prepared via Method B using N-(4-piperidin-l-ylbenzyl)propan-2- amine.
  • This compound was prepared via Method B using propyl-(tetrahydro-furan-2-ylmethyl)- amine.
  • This compound was prepared via Method I using l-pyridin-2-yl-piperazine.
  • This compound was prepared via Method B using (4-fluoro-benzyl)-(2-methoxy-l- methyl-ethyl)-amine hydrochloride.
  • This compound was prepared via Method B using l-piperidin-4-yl-lH-benzotriazole.
  • This compound was prepared via Method I using methyl-phenethyl-amine.
  • This compound was prepared via Method I using methyl-(4-pyridin-2-yl-benzyl)-amine.
  • This compound was prepared via Method I using methyl-pyridin-3-ylmethyl-amine.
  • This compound was prepared via Method F using 2-(4-benzyloxy-phenyl)-4,4,5,5- tetramethyl- [ 1 ,3 ,2] dioxaborolane.
  • This compound was prepared via Method L using morpholine.
  • This compound was prepared via Method L using N-methyl morpholine.
  • This compound was prepared via Method E using C-(l,5-dimethyl-lH-pyrazol-3-yl)- methylamine.
  • This compound was prepared via Method L using pirrolidine.
  • Sy n th es is of 5-[4-(4,4,5,5-Tetramethyl-[l,3,2Jdioxaborolan-2-yl)-phenoxymethylJ- pyridine-2-carbonitrile
  • This compound was prepared via Method D using 1 -cyano-cyclopropanecarbonyl chloride.
  • This compound was prepared via Method D using phenyl-methanesulfonyl chloride.
  • This compound was prepared via Method C using the mesylate derivative of 6- hydroxymethyl-nicotinic acid methyl ester.
  • This compound was prepared via Method D using cyclopropanesulfonyl chloride.
  • This compound was prepared via Method C using 3-chloromethyl-pyridine 1 -oxide.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Pulmonology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Transplantation (AREA)
  • Otolaryngology (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

[1,2,4]Triazolo[1,5-a]pyridine compounds are disclosed that have a formula represented by the formula (I). The compounds may be prepared as pharmaceutical compositions, and may be used for the prevention and treatment of a variety of conditions in mammals including humans, including by way of non- limiting 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. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6, transplantation rejection (e.g. organ transplant rejection) and proliferative diseases.

Description

NOVEL COMPOUNDS USEFUL FOR THE TREATMENT OF DEGENERATIVE AND
INFLAMMATORY DISEASES
FIELD OF THE INVENTION
[0001] 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.
[0002] Janus kinases (JAKs) are cytoplasmic tyrosine kinases that transduce cytokine signaling from membrane receptors to STAT transcription factors. Four JAK family members are described, JAKl, JAK2, JAK3 and TYK2. 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).
[0003] The combination of genetic models and small molecule JAK inhibitor research revealed the therapeutic potential of several JAKs. 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/). [0004] 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)).
[0005] 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.
BACKGROUND OF THE INVENTION
[0006] 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. In cartilage, 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.
[0007] One of the functional roles of cartilage in the joint is to allow bones to articulate on each other smoothly. Loss of articular cartilage, therefore, causes the bones to rub against each other leading to pain and loss of mobility. The degradation of cartilage can have various causes. In inflammatory arthritides, as rheumatoid arthritis for example, 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.
[0008] The degeneration of cartilage is the hallmark of various diseases, among which rheumatoid arthritis and osteoarthritis are the most prominent. Rheumatoid arthritis (RA) is a chronic joint degenerative disease, characterized by inflammation and destruction of the joint structures. When the disease is unchecked, it leads to substantial disability and pain due to loss of joint functionality and even premature death. The aim of an RA therapy, therefore, is not to slow down the disease but to attain remission in order to stop the joint destruction. Besides the severity of the disease outcome, the high prevalence of RA (~ 0.8% of the adults are affected worldwide) means a high socio-economic impact. (For reviews on RA, we refer to Smolen and Steiner (2003); Lee and Weinblatt (2001); Choy and Panayi (2001); O'Dell (2004) and Firestein (2003)).
[0009] Osteoarthritis (also referred to as OA, or wear-and-tear arthritis) is the most common form of arthritis and is characterized by loss of articular cartilage, often associated with hypertrophy of the bone and pain. 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. There are further levels of degeneration and destruction, which affect the deep and the calcified cartilage layers that border with the subchondral bone. For an extensive review on osteoarthritis, we refer to Wieland et al., 2005.
[0010] The clinical manifestations of the development of the osteoarthritis condition are: increased volume of the joint, pain, crepitation and functional disability that lead to pain and reduced mobility of the joints. When disease further develops, pain at rest emerges. If the condition persists without correction and/or therapy, the joint is destroyed leading to disability. Replacement surgery with total prosthesis is then required. [0011] Therapeutic methods for the correction of the articular cartilage lesions that appear during the osteoarthritic disease have been developed, but so far none of them have been able to mediate the regeneration of articular cartilage in situ and in vivo.
[0012] 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, 2006). Taken together, no disease modifying osteoarthritic drugs are available.
[0013] In severe cases, joint replacement may be necessary. This is especially true for hips and knees. If a joint is extremely painful and cannot be replaced, it may be fused. This procedure stops the pain, but results in the permanent loss of joint function, making walking and bending difficult. [0014] Another possible treatment is the transplantation of cultured autologous chondrocytes.
Here, 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.
[0015] 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. [0016] Other reported methods include application of tendinous, periosteal, fascial, muscular or perichondral grafts; implantation of fibrin or cultured chondrocytes; implantation of synthetic matrices, such as collagen, carbon fiber; administration of electromagnetic fields. All of these have reported minimal and incomplete effects, resulting in a poor quality tissue that can neither support the weighted load nor allow the restoration of an articular function with normal movement.
[0017] 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. Insulin-like growth factor-I (IGF-I) is the predominant anabolic growth factor in synovial fluid and stimulates the synthesis of both proteoglycans and collagen. It has also been shown that members of the bone morphogenetic protein (BMP) family, notably BMP2, BMP4, BMP6, and BMP7, and members of the human transforming growth factor-β (TGF-β) family can induce chondrocyte anabolic stimulation (Chubinskaya and Kuettner, 2003). A compound has recently been identified that induces anabolic stimulation of chondrocytes (US 6,500,854; EP 1 391 211). However, most of these compounds show severe side effects and, consequently, there is a strong need for compounds that stimulate chondrocyte differentiation without these side effects. [0018] Vandeghinste et al. (WO 2005/124342) discovered JAKl as a target whose inhibition might have therapeutic relevance for several diseases including OA. 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). 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. Moreover, 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 ef α/., 1998).
[0019] Various groups have implicated JAK-STAT signaling in chondrocyte biology. Li et al
(2001) showed that Oncostatin M induces MMP and TIMP3 gene expression in primary chondrocytes by activation of JAK/STAT and MAPK signaling pathways. Osaki et al. (2003) showed that interferon- gamma mediated inhibition of collagen II in chondrocytes involves JAK-STAT signaling. ILl -beta induces cartilage catabolism by reducing the expression of matrix components, and by inducing the expression of collagenases and inducible nitric oxide synthase (NOS2), which mediates the production of nitric oxide (NO). Otero et al., (2005) showed that leptin and ILl -beta synergistically induced NO production or expression of NOS2 mRNA in chondrocytes, and that that was blocked by a JAK inhibitor. Legendre et al. (2003) showed that IL6/IL6Receptor induced downregulation of cartilage- specific matrix genes collagen II, aggrecan core and link protein in bovine articular chondrocytes, and that this was mediated by JAK/STAT signaling. Therefore, these observations suggest a role for JAK kinase activity in cartilage homeostasis and therapeutic opportunities for JAK kinase inhibitors. [0020] 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. 44(10):2497-506; Xiang et al, 2008, "Identification of somatic JAKl mutations in patients with acute myeloid leukemia" Blood First Edition Paper, prepublished online December 26, 2007; DOI 10.1182/blood-2007-05-090308) and acute lymphoblastic leukemia (Mullighan et al, 2009) or solid tumours e.g. uterine leiomyosarcoma (Constantinescu et al, 2007, Trends in Biochemical Sciences 33(3): 122-131), prostate cancer (Tarn et al, 2007, British Journal of Cancer, 97, 378 - 383) These results indicate that inhibitors of JAK, in particular of JAKl and/or JAK2, may also have utility in the treatment of cancers (leukaemias and solid tumours e.g. uterine leiomyosarcoma, prostate cancer).
[0021] In addition, Castleman's disease, multiple myeloma, mesangial proliferative glomerulonephritis, psoriasis, and Kaposi's sarcoma are likely due to hypersecretion of the cytokine IL- 6, whose biological effects are mediated by intracellular JAK-STAT signaling (Tetsuji Naka, Norihiro Nishimoto and Tadamitsu Kishimoto, Arthritis Res 2002, 4 (suppl 3):S233-S242). This result shows that inhibitor of JAK, may also find utility in the treatment of said diseases.
[0022] 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.
[0023] 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 diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular 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.
SUMMARY OF THE INVENTION
[0024] 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 diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular 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.
[0025] Accordingly, in a first aspect of the invention, l,2,4-triazolo[l,5-a]pyridine compounds are disclosed having a Formula (I):
Figure imgf000007_0001
wherein
CyI is selected from aryl and heteroaryl;
Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)-, - S(O)2-, - N(R4a)CO-, -CH2-N(R4a)- or - N(R4a)SO2-; each R1 is independently selected from Ci-Ce alkyl, substituted Ci-Cβ alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted CpC6 alkoxy, substituted or unsubstituted amido, substituted or unsubstituted amino, substituted sulfinyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4- 7 membered heterocycloalkyl, halo, and hydroxyl; each R3a is independently selected from CpC6 alkyl, substituted CpC6 alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted CpC6 alkoxy, substituted or unsubstituted amido, alkoxycarbonyl, substituted alkoxycarbonyl, arylalkyloxy, substituted arylalkyloxy, substituted or unsubstituted amino, aryl, substituted aryl, arylalkyl, substituted sulfanyl, substituted sulfinyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, halo, substituted or unsubstituted heteroaryl, hydroxyl, nitro, and thiol; R a is selected from substituted or unsubstituted Ci-Ce alkyl and substituted or unsubstituted C3-C7 cycloalkyl;
R3b is independently selected from substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5-10 membered heteroaryl; or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from substituted Ci-C6 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5- 10 membered heteroaryl; each R4a, R4b and R4c is independently selected from H, Ci-C6 alkyl, substituted Ci-C6 alkyl, C3-C7 cycloalkyl, or substituted C3-C7 cycloalkyl, substituted or unsubstituted acyl; ml is 0, 1, or 2; m2 is 0, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that i) when Ll is -O-, -N(R4a)-, -CH2-N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1, 2, 3, or 4; ii) when CyI is Ph, Ll is a bond, nl is 0, and R3b is -OR3c, then R3c is other than Me or
CF3; or a pharmaceutically acceptable salts or solvates thereof, or solvates of the pharmaceutically acceptable salts.
[0026] In a further aspect, the present invention l,2,4-triazolo[l,5-a]pyridine compounds according to Formula I are disclosed that are capable of capable of modulating the activity of JAK in vivo.
[0027] In a further aspect, the present invention provides pharmaceutical compositions comprising a compound of the invention, and a pharmaceutical carrier, excipient or diluent. In this aspect of the invention, the pharmaceutical composition can comprise one or more of the compounds described herein. Moreover, the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein, are all pharmaceutically acceptable as prepared and used.
[0028] In a further aspect of the invention, 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. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection), which method comprises administering an effective amount of one or more of the pharmaceutical compositions or compounds described herein. Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer). In a particular embodiment 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 described herein.
[0029] In a further aspect, the present invention provides compounds according to Formula I or a pharmaceutically acceptable salt thereof in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases.
[0030] In a further aspect, 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.
[0031] In a further aspect, 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. chronic cardiac failure), diseases involving impairment of cartilage turnover (e.g diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer). In a specific embodiment, the condition is 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.
[0032] In a further aspect, 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.
[0033] In yet another method of treatment aspect, 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.
[0034] In a further aspect, 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.
[0035] In additional aspects, this invention provides methods for synthesizing the compounds of the invention, with representative synthetic protocols and pathways disclosed later on herein.
[0036] Accordingly, it is a principal object of this invention to provide a novel series of compounds, which can modify the activity of JAK and thus prevent or treat any maladies that may be causally related thereto.
[0037] It is further an object of this invention to provide a series of compounds that can treat or alleviate maladies or symptoms of same, such as cartilage and/or bone degradation and related inflammation, and joint diseases, that may be causally related to the activity of JAK.
[0038] 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 diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer). In a specific embodiment the condition is selected from inflammation, 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), and organ transplant rejection; and cartilage, bone and/or joint degradation or degeneration, such as osteoarthritis or cancers (e.g. solid tumours or leukaemias). [0039] Other objects and advantages will become apparent to those skilled in the art from a consideration of the ensuing detailed description.
DETAILED DESCRIPTION OF THE INVENTION Definitions
[0040] The following terms are intended to have the meanings presented therewith below and are useful in understanding the description and intended scope of the present invention.
[0041] When describing the invention, which may include compounds, pharmaceutical compositions containing such compounds and methods of using such compounds and compositions, the following terms, if present, have the following meanings unless otherwise indicated. It should also be understood that when described herein any of the moieties defined forth below may be substituted with a variety of substituents, and that the respective definitions are intended to include such substituted moieties within their scope as set out below. Unless otherwise stated, the term "substituted" is to be defined as set out below. It should be further understood that the terms "groups" and "radicals" can be considered interchangeable when used herein.
[0042] The articles "a" and "an" may be used herein to refer to one or to more than one (i.e. at least one) of the grammatical objects of the article. By way of example "an analogue" means one analogue or more than one analogue.
[0043] 'Acyl' refers to a radical -C(O)R20, where R20 is hydrogen, C1-C8 alkyl, C3-Ci0 cycloalkyl, C3-Ci0 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-C8 alkyl, -C(O)-(CH2MC6-C10 aryl), -C(O)-(CH2)t(5-
10 membered heteroaryl), -C(O)-(CH2)t(C3-Ci0 cycloalkyl), and -C(O)-(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4.
[0044] 'Substituted Acyl' refers to a radical -C(O)R21, wherein R21 is independently • CpC8 alkyl, substituted with halo or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of which is substituted with unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0045] 'Acylamino' refers to a radical -NR22C(O)R23, where R22 is hydrogen, CpC8 alkyl, C3-
Cio cycloalkyl, 4-10 membered heterocycloalkyl, C6-CiO aryl, arylalkyl, 5-10 memberd heteroaryl or heteroarylalkyl and R is hydrogen, CpC8 alkyl, C3-CiO cycloalkyl, 4-10 membered heterocycloalkyl, C6-CiO 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 -NR21 C(O)-Ci-C8 alkyl, -NR21'C(O)-(CH2)t(C6-Ci0 aryl), -NR21 C(O)-(CH2)t(5-10 membered heteroaryl), -NR21'C(O)-(CH2)t(C3-Ci0 cycloalkyl), and -NR21 C(O)-(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R21 independently represents H or CpC8 alkyl.
[0046] 'Substituted Acylamino' refers to a radical -NR24C(O)R25, wherein:
R24 is independently
• H, CpC8 alkyl, substituted with halo or hydroxy; or
• C3-CiO cycloalkyl, 4-10 membered heterocycloalkyl, C6-CiO aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of which is substituted with unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy; and
R 5 is independently
• H, CpC8 alkyl, substituted with halo or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of which is substituted with unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxyl; provided at least one of R24 and R25 is other than H.
[0047] 'Alkoxy' refers to the group -OR26 where R26 is CpC8 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.
[0048] '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, Cβ-Cio aryl, -O- aryl, carboxyl, cyano, C3-C10 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 -0-(CH2)t(C6-Cio aryl), -O-(CH2)t(5-10 membered heteroaryl), -0-(CH2)t(C3-Cio cycloalkyl), and -O-(CH2)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 C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. Particular exemplary 'substituted alkoxy' groups are OCF3, OCH2CF3, OCH2Ph, OCH2- cyclopropyl, OCH2CH2OH, OCH2CH2NMe2.
[0049] 'Alkoxycarbonyl' refers to a radical -C(O)-OR27 where R27 represents an Ci-C8 alkyl,
C3-CiO cycloalkyl, C3-CiO cycloalkylalkyl, 4-10 membered heterocycloalkylalkyl, aralkyl, or 5-10 membered heteroarylalkyl as defined herein. Exemplary "alkoxycarbonyl" groups are C(O)O-CpCg alkyl, -C(O)O-(CH2MC6-C10 aryl), -C(O)O-(CH2)t(5-10 membered heteroaryl), -C(O)O-(CH2MC3-C10 cycloalkyl), and -C(O)O-(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 1 to 4. [0050] 'Substituted Alkoxycarbonyl' refers to a radical -C(O)-OR28 where R28 represents:
• CpC8 alkyl, C3-Ci0 cycloalkyl, C3-Ci0 cycloalkylalkyl, or 4-10 membered heterocycloalkylalkyl, each of which is substituted with halo, substituted or unsubstituted amino, or hydroxy; or
• Cβ-Cio aralkyl, or 5-10 membered heteroarylalkyl, each of which is substituted with unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxyl.
[0051] '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.
[0052] '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)R20), alkoxy, alkoxycarbonyl, alkoxycarbonylamino (-NR -alkoxycarbonyl or -NH- C(O)-OR27), amino, substituted amino, aminocarbonyl (carbamoyl or amido or -C(O)-NR 2), aminocarbonylamino (-NR -C(O)-NR 2), aminocarbonyloxy (-0-C(O)-NR 2), aminosulfonyl, sulfonylamino, aryl, -O-aryl, azido, carboxyl, cyano, cycloalkyl, halogen, hydroxy, heteroaryl, nitro, thiol, -S-alkyl, -S-aryl, -S(O)-alkyl,-S(O)-aryl, -S(O)2-alkyl, and -S(O)2-aryl. In a particular embodiment 'substituted alkyl' refers to a CpCg alkyl group substituted with halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR"'SO2R", -SO2NR"R"', -C(O)R", -C(O)OR", -OC(O)R", - NR"'C(O)R", -C(O)NR R", -NR"R ", or -(CR"'R"")mOR"; wherein each R" is independently selected from H, Ci-C8 alkyl, -(CH2MC6-C10 aryl), -(CH2)t(5-10 membered heteroaryl), -(CH2MC3-C10 cycloalkyl), and -(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from O to 4 and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted C1-C4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. Each of R and R independently represents H or CpCg alkyl. [0053] 'Amino' refers to the radical -NH2.
[0054] '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 )2 where each R is independently selected from:
• hydrogen, CpCg alkyl, C6-Ci0 aryl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, or C3-Ci0 cycloalkyl; or
• CpCg alkyl, substituted with halo or hydroxy; or
• -(CH2MC6-Ci0 aryl), -(CH2)t(5-10 membered heteroaryl), -(CH2MC3-Ci0 cycloalkyl) or - (CH2)t(4-10 membered heterocycloalkyl) wherein t is an integer between 0 and 8, each of which is substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy; or
• both R33 groups are joined to form an alkylene group.
When both R groups are hydrogen, -N(R )2 is an amino group. Exemplary 'substituted amino' groups are -NR33'-CpC8 alkyl, -NR33'-(CH2)t(C6-Ci0 aryl), -NR33'-(CH2)t(5-10 membered heteroaryl), -NR33'- (CH2MC3-Ci0 cycloalkyl), and -NR -(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R33 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 unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. For the avoidance of doubt the term "substituted amino" includes the groups alkylamino, substituted alkylamino, dialkylamino and substituted dialkylamino as defined below.
[0055] 'Alkylamino' refers to the group -NHR34, wherein R34 is CpC8 alkyl.
[0056] 'Substituted Alkylamino' refers to the group -NHR35, wherein R35 is CpC8 alkyl; and the alkyl group is substituted with halo, substituted or unsubstituted amino, hydroxy, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, 5-10 membered heteroaryl, aralkyl or heteroaralkyl; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted
CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0057] 'Dialkylamino' refers to the group -NR42R43, wherein each of R42 and R43 are independently selected from CpCg alkyl.
[0058] 'Substituted Dialkylamino' refers to the group -NR44R45, wherein each of R44 and R45 are independently selected from CpCg alkyl; and the alkyl group is independently substituted with halo, hydroxy, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, 5-10 membered heteroaryl, aralkyl or heteroaralkyl; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted
Cp4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0059] 'Aminosulfonyl' or 'Sulfonamide' refers to the radical -S(O2)NH2.
[0060] 'Substituted aminosulfonyl' or 'substituted sulfonamide' refers to a radical such as -
S(O2)N(R48)2 wherein each R48 is independently selected from:
• H, CpCg alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, and heteroaralkyl; or
• CpCg alkyl substituted with halo or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy; provided that at least one R48 is other than H.
[0061] Exemplary 'substituted aminosulfonyl' or 'substituted sulfonamide' groups are -
S(O2)N(R48')-CpCg alkyl, -S(O2)N(R48')-(CH2)t(C6-Ci0 aryl), -S(O2)N(R48')-(CH2)t(5-10 membered heteroaryl), -S(O2)N(R48')-(CH2)t(C3-Ci0 cycloalkyl), and -S(O2)N(R48')-(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4; each R48 independently represents H or CpCg alkyl; and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. [0062] '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. In particular aryl refers to an aromatic ring structure, mono-cyclic or poly-cyclic that includes from 5 to 12 ring members, more usually 6 to 10. Where the 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, triphenylene and trinaphthalene. Particularly aryl groups include phenyl, naphthyl, indenyl, and tetrahydronaphthyl. [0063] '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. Particularly, 'Substituted Aryl' refers to an aryl group substituted with one or more of groups selected from halo, CpC8 alkyl, CpC8 haloalkyl, CpC8 haloalkoxy, cyano, hydroxy, CpC8 alkoxy, and amino. [0064] Examples of representative substituted aryls include the following
Figure imgf000016_0001
[0065] In these formulae one of R49 and R50 may be hydrogen and at least one of R49 and R50 is each independently selected from CpC8 alkyl, 4-10 membered heterocycloalkyl, CpC8 alkoxy, hetero- O-aryl, alkylamino, NR51COR52, NR51SOR52 NR51SO2R52, COOalkyl, COOaryl, CONR51R52, CONR51OR52, NR51R52, SO2NR51R52, S-alkyl, SOalkyl, S02alkyl, Saryl, SOaryl, S02aryl; or R49 and R50 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. R51, and R52 are independently hydrogen, CpC8 alkyl, CpC4 haloalkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, substituted aryl, 5-10 membered heteroaryl.
[0066] 'Arylalkyloxy' refers to an -O-alkylaryl radical where alkylaryl is as defined herein.
[0067] 'Substituted Arylalkyloxy' refers to an -O-alkylaryl radical where alkylaryl is as defined herein; and any aryl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, cyano, unsubstituted CpC4 alkoxy, unsubstituted Cp4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. [0068] 'Azido' refers to the radical -N3.
[0069] 'Amido' refers to the radical -C(O)NH2.
[0070] 'Substituted amido' refers to the radical -C(O)N(R53)2 wherein each R53 is independently
• H, CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, and heteroaralkyl; or
• CpC8 alkyl substituted with halo or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, each of which is substituted by unsubstituted CpC4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxy; provided that at least one R53 is other than H.
Exemplary 'Substituted Amido ' groups are -C(O) NR53'-CrC8 alkyl, -C(0)NR53'-(CH2)t(C6-Cio aryl), - C(O)N53'-(CH2)t(5-10 membered heteroaryl), -C(0)NR53'-(CH2)t(C3-Cio cycloalkyl), and -C(O)NR53'- (CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4, each R53 independently represents H or CpCg alkyl and any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. [0071] 'Carboxy' refers to the radical -C(O)OH.
[0072] '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.
[0073] '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. [0074] 'Cyano' refers to the radical -CN.
[0075] 'Halo' or 'halogen' refers to fluoro (F), chloro (Cl), bromo (Br) and iodo (I). Particular halo groups are either fluoro or chloro.
[0076] '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. [0077] '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. Typically 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. In one embodiment, 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. Particular examples of bicyclic heteroaryl groups containing a five membered ring fused to another five membered ring include but are not limited to imidazothiazole and imidazoimidazole. Particular examples of 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. Particular examples of 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. Particular heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine.
[0078] Examples of representative aryl having hetero atoms containing substitution include the following:
Figure imgf000018_0001
wherein each W is selected from C(R54)2, NR54, O and S; and each Y is selected from carbonyl, NR54, O and S; and R54 is independently hydrogen, CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, and 5-10 membered heteroaryl. [0079] Examples of representative heteroaryls include the following:
Figure imgf000018_0002
Figure imgf000018_0003
wherein each Y is selected from carbonyl, N, NR55, O and S; and R55 is independently hydrogen, CpC8 alkyl, C3-C10 cycloalkyl, 4-10 membered heterocycloalkyl, Cβ-Cio aryl, and 5-10 membered heteroaryl. [0080] As used herein, 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. Examples of 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. 4-tetrahydro pyranyl), imidazoline, imidazolidinone, oxazoline, thiazoline, 2-pyrazoline, pyrazolidine, piperazine, and N-alkyl piperazines such as N-methyl piperazine. Further examples include thiomorpholine and its S-oxide and S,S-dioxide (particularly thiomorpholine). Still further examples include azetidine, piperidone, piperazone, and N-alkyl piperidines such as N-methyl piperidine. Particular examples of heterocycloalkyl groups are shown in the following illustrative examples:
Figure imgf000019_0001
wherein each W is selected from CR56, C(R56)2, NR56, O and S; and each Y is selected from NR56, O and S; and R56 is independently hydrogen, CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, Cβ-Cio 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)R20), alkoxy, alkoxycarbonyl, alkoxycarbonylamino (-NR -alkoxycarbonyl or -NH-C(O)-OR27), amino, substituted amino, aminocarbonyl (amido or -C(O)-NR 2), aminocarbonylamino (-NR -C(O)- NR 2), aminocarbonyloxy (-O-C(O)-NR 2), aminosulfonyl, sulfonylamino, aryl, -O-aryl, azido, carboxyl, cyano, cycloalkyl, halogen, hydroxy, nitro, thiol, -S-alkyl, -S-aryl, -S(O)-alkyl,-S(O)-aryl, -S(O)2-alkyl, and -S(0)2-aryl. Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivatives.
[0081] 'Hydroxy' refers to the radical -OH.
[0082] 'Nitro' refers to the radical -NO2. [0083] 'Substituted' refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s). Typical substituents may be selected from the group consisting of: halogen, -R57, -O\ =0, -OR57, -SR57, -S", =S, -NR57R58, =NR57, -CCl3, -CF3, -CN, -OCN, -SCN, -NO, - NO2, =N2, -N3, -S(O)2O", -S(O)2OH, -S(O)2R57, -OS(O2)O", -OS(O)2R57, -P(O)(O )2, -P(O)(OR57)(O"), -OP(O)(OR57XOR58), -C(O)R57, -C(S)R57, -C(O)OR57, -C(O)NR57R58, -C(O)O", -C(S)OR57, - NR59C(O)NR57R58, -NR59C(S)NR57R58, -NR60C(NR59)NR57R58 and -C(NR59)NR57R58; wherein each R57, R58, R59 and R60 are independently:
• hydrogen, CpC8 alkyl, C6-Ci0 aryl, arylalkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, 5-10 membered heteroaryl, heteroarylalkyl; or
• CpCg alkyl substituted with halo or hydroxy; or
• C6-Ci0 aryl, 5-10 membered heteroaryl, C6-Ci0 cycloalkyl or 4-10 membered heterocycloalkyl substituted by unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
In a particular embodiment, substituted groups are substituted with one or more substituents, particularly with 1 to 3 substituents, in particular with one substituent group.
In a further particular embodiment the substituent group or groups are selected from: halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR"'SO2R", -SO2NR R", -C(O)R", -C(O)OR", -OC(O)R", - NR'"C(O)R", -C(O)NR R", -NR"R ", -(CR'"R'")mOR'", wherein, each R" is independently selected from H, CpC8 alkyl, -(CH2)t(C6-Ci0 aryl), -(CH2)t(5-10 membered heteroaryl), -(CH2)t(C3-Ci0 cycloalkyl), and -(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from O to 4; and
• any alkyl groups present, may themselves be substituted by halo or hydroxy; and
• any aryl, heteroaryl, cycloalkyl or heterocycloalkyl groups present, may themselves be substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. Each R independently represents H or CpC6alkyl.
[0084] 'Substituted sulfanyl' refers to the group -SR61, wherein R61 is selected from:
• CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5- 10 membered heteroaryl, and heteroaralkyl; or
• CpC8 alkyl substituted with halo, substituted or unsubstituted amino, or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, each of which is substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. [0085] Exemplary 'substituted sulfanyl' groups are -S-(CpC8 alkyl) and -S-(C3-Ci0 cycloalkyl), -S-(CH2)t(C6-Cio aryl), -S-(CH2)t(5-10 membered heteroaryl), -S-(CH2)t(C3-Ci0 cycloalkyl), and -S-(CH2)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 C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxy. The term 'substituted sulfanyl' includes the groups 'alkylsulfanyl' or 'alkylthio', 'substituted alkylthio' or 'substituted alkylsulfanyl', 'cycloalkylsulfanyl' or 'cycloalkylthio', 'substituted cycloalkylsulfanyl' or 'substituted cycloalkylthio', 'arylsulfanyl' or 'arylthio' and 'heteroarylsulfanyl' or 'heteroarylthio' as defined below. [0086] Substituted sulfinyl' refers to the group -S(O)R68, wherein R68 is selected from:
• CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5- 10 membered heteroaryl, and heteroaralkyl; or
• CpC8 alkyl substituted with halo, substituted or unsubstituted amino, or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, substituted by unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0087] Exemplary 'substituted sulfinyl' groups are -S(O)-(CpC8 alkyl) and -S(O)-(C3-Ci0 cycloalkyl), -S(O)-(CH2MC6-C10 aryl), -S(O)-(CH2)t(5-10 membered heteroaryl), -S(O)-(CH2MC3-C10 cycloalkyl), and -S(O)-(CH2)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 CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. The term substituted sulfinyl includes the groups 'alkylsulfinyl', 'substituted alkylsulfinyP, 'cycloalkylsulfinyl', 'substituted cycloalkylsulfinyP, 'arylsulfinyP and 'heteroarylsulfmyP as defined herein. [0088] 'Substituted sulfonyP refers to the group -S(O)2R75, wherein R75 is selected from:
• CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, and heteroaralkyl; or
• CpC8 alkyl substituted with halo, substituted or unsubstituted amino, or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, each of which is substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0089] Exemplary 'substituted sulfonyP groups are -S(O)2-(CpC8 alkyl) and -S(O)2-(C3-Ci0 cycloalkyl), -S(O)2-(CH2MC6-C10 aryl), -S(O)2-(CH2)t(5-10 membered heteroaryl), -S(O)2-(CH2MC3- Ci0 cycloalkyl), and -S(O)2-(CH2)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 CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. The term substituted sulfonyl includes the groups alkylsulfonyl, substituted alkylsulfonyl, cycloalkylsulfonyl, substituted cycloalkylsulfonyl, arylsulfonyl and heteroarylsulfonyl.
[0090] 'Sulfo' or 'sulfonic acid' refers to a radical such as -SO3H.
[0091] 'Substituted sulfo' or 'sulfonic acid ester' refers to the group -S(O)2OR82, wherein R82 is selected from:
• CpC8 alkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, and heteroaralkyl; or
• CpC8 alkyl substituted with halo, substituted or unsubstituted amino, or hydroxy; or
• C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, each of which is substituted by unsubstituted CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy.
[0092] Exemplary 'Substituted sulfo' or 'sulfonic acid ester' groups are -S(O)2-O-(CpC8 alkyl) and -S(O)2-O-(C3-Ci0 cycloalkyl), -S(O)2-O-(CH2MC6-C10 aryl), -S(O)2-O-(CH2)t(5-10 membered heteroaryl), -S(O)2-O-(CH2X(C3-Ci0 cycloalkyl), and -S(O)2-O-(CH2)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 CpC4 alkyl, halo, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 hydroxyalkyl, or unsubstituted CpC4 haloalkoxy or hydroxy. [0093] 'Thiol' refers to the group -SH.
[0094] One having ordinary skill in the art of organic synthesis will recognize that the maximum number of heteroatoms in a stable, chemically feasible heterocyclic ring, whether it is aromatic or non aromatic, is determined by the size of the ring, the degree of unsaturation and the valence of the heteroatoms. In general, a heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.
[0095] 'Pharmaceutically acceptable' means approved or approvable by a regulatory agency of the Federal or a state government or the corresponding agency in countries other than the United States, or that is listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly, in humans.
[0096] '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. In particular, such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts. Specifically, 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-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-l-carboxylic acid, glucoheptonic acid, 3- phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine and the like. 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. The term "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.
[0097] 'Pharmaceutically acceptable vehicle' refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
[0098] '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. [0099] 'Solvate' refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. This physical association includes hydrogen bonding. Conventional 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.
[00100] 'Subject' includes humans. The terms 'human', 'patient' and 'subject' are used interchangeably herein. [00101] '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.
[00102] '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.
[00103] 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 anti-malarial 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.
[00104] '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). In another embodiment 'treating' or 'treatment' refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, '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. In a further embodiment, "treating" or "treatment" relates to slowing the progression of the disease.
[00105] As used herein the term '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. Partcicularly the term refers to rheumatoid arthritis, osteoarthritis, allergic airway disease (e.g. asthma) and inflammatory bowel diseases.
[00106] As used herein the terms 'condition(s) involving an immune response' or 'autoimmune diseases' are used interchangeably and refer to refers to the group of diseases including 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 dermatitises, inflammatory bowel disease (e.g. Crohn's disease and ulcerative colitis), atherosclerosis and amyotrophic lateral sclerosis. Particularly the term refers to COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and inflammatory bowel disease.
[00107] As used herein 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.
[00108] As used herein the term 'proliferative disease(s)' refers to conditions such as cancer
(e.g. uterine leiomyosarcoma or prostate cancer), myeloproliferative disorders, named in particular Re JAK2 activating mutations (polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis), leukemia (e.g. acute myeloid leukaemia and acute lymphoblastic leukemia), multiple myeloma, psoriasis, restenosis, sclerodermitis or fibrosis. In particular the term refers to cancer, leukemia, multiple myeloma and psoriasis.
[00109] As used herein, 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. As used herein the term 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.
[00110] As used herein the term '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. In particular the term leukemia refers to acute myeloid leukaemia (AML) and acute lymphoblastic leukemia (ALL).
[00111] As used herein the term 'diseases involving impairment of cartilage turnover' or
"diseases involving the anabolic stimulation of chondrocytes" includes conditions such as 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. [00112] As used herein 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. [00113] As used herein 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.
[00114] 'Compound(s) of the invention', and equivalent expressions, are meant to embrace compounds of the Formula(e) as hereinbefore described, which expression includes the prodrugs, the pharmaceutically acceptable salts, and the solvates, e.g., hydrates, and the solvates of the pharmaceutically acceptable salts where the context so permits. Similarly, reference to intermediates, whether or not they themselves are claimed, is meant to embrace their salts, and solvates, where the context so permits.
[00115] When ranges are referred to herein, for example but without limitation, CpCg alkyl, the citation of a range should be considered a representation of each member of said range. [00116] Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but in the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). 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, C2-Cg alkenyl, aryl, C7-C12 substituted aryl, and C7-C12 arylalkyl esters of the compounds of the invention.
[00117] As used herein, 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. For example, an 'isotopic variant' of a compound can contain one or more non-radioactive isotopes, such as for example, deuterium (2H or D), carbon- 13 (13C), nitrogen- 15 (15N), or the like. It will be understood that, in a compound where such isotopic substitution is made, the following atoms, where present, may vary, so that for example, any hydrogen may be 2H/D, any carbon may be 13C, or any nitrogen may be 15N, and that the presence and placement of such atoms may be determined within the skill of the art. Likewise, 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. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as 11C, 18F, 15O and 13N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. [00118] All isotopic variants of the compounds provided herein, radioactive or not, are intended to be encompassed within the scope of the invention.
[00119] It is also to be understood that compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed 'isomers'. Isomers that differ in the arrangement of their atoms in space are termed
'stereoisomers'.
[00120] Stereoisomers that are not mirror images of one another are termed 'diastereomers' and those that are non-superimposable mirror images of each other are termed 'enantiomers'. When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. 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'.
[00121] '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 π electrons and an atom (usually H). For example, 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 phenylnitromethane, that are likewise formed by treatment with acid or base.
[00122] Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.
[00123] 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.
[00124] Unless indicated otherwise, 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 COMPOUNDS
[00125] 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 diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer). In particular 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.
[00126] Accordingly, in a first aspect of the invention, l,2,4-triazolo[l,5-a]pyridine compounds are disclosed having a Formula (I):
Figure imgf000028_0001
wherein
CyI is selected from aryl and heteroaryl;
Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)- , -S(O)2-, - N(R4a)CO-, -CH2-N(R4a)- or - N(R4a)SO2-; each R1 is independently selected from CpC6 alkyl, substituted CpC6 alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted Ci-Ce alkoxy, substituted or unsubstituted amido, substituted or unsubstituted amino, substituted sulfmyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R3a is independently selected from CpC6 alkyl, substituted CpC6 alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted Ci-Cβ alkoxy, substituted or unsubstituted amido, alkoxycarbonyl, substituted alkoxycarbonyl, arylalkyloxy, substituted arylalkyloxy, substituted or unsubstituted amino, aryl, substituted aryl, arylalkyl, substituted sulfanyl, substituted sulfinyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, halo, substituted or unsubstituted heteroaryl, hydroxyl, nitro, and thiol; R2a is selected from substituted or unsubstituted CpC6 alkyl and substituted or unsubstituted C3-
C7 cycloalkyl;
R3b is independently selected from substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5-10 membered heteroaryl; or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from substituted Cr C6 alkyl, substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5- 10 membered heteroaryl; each R4a, R4b and R4c is independently selected from H, Ci-C6 alkyl, substituted Ci-C6 alkyl, C3-
C7 cycloalkyl, or substituted C3-C7 cycloalkyl, substituted or unsubstituted acyl; ml is 0, 1, or 2; m2 is 0, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that i) when Ll is -O-, -N(R4a)-, -CH2-N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1, 2, 3, or 4; ii) when CyI is Ph, Ll is a bond, nl is 0, and R3b is -OR3c, then R3c is other than Me or
CF3; or pharmaceutically acceptable salts or solvates thereof or a solvates of pharmaceutically acceptable salts.
[00127] In a further aspect, the present invention provides compounds according to Formula I or a pharmaceutically acceptable salt thereof in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases.
[00128] In a further embodiment, the compound is according to Formula (I) above wherein:
CyI is selected from aryl and heteroaryl; Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)-
, -S(O)2-, - N(R4a)CO-, -CH2-N(R4a)-or - N(R4a)SO2-; each R1 is independently selected from unsubstituted CpC6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted Ci-C6 alkoxy, unsubstituted amido, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from unsubstituted Ci-Ce alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted CpC6 alkoxy, unsubstituted amido, unsubstituted alkoxycarbonyl, unsubstituted arylalkyloxy, unsubstituted amino, unsubstituted aryl, unsubstituted arylalkyl, aminosulfonyl (which aminosulfonyl may be substituted with unsubstituted C1-C4 alkyl), sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, unsubstituted heteroaryl, hydroxyl, nitro, and thiol;
R a is selected from unsubstituted CpC6 alkyl and unsubstituted C3-C7 cycloalkyl;
R3b is independently selected from aryl (which aryl may be substituted with halo, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted C1-C4 alkyl, unsubstituted C1-C4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 haloalkoxy, unsubstituted 5-7- membered heteroaryl, unsubstituted acylamino, unsubstituted amino, cyano, -(CH2)i_4- CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted with cyano), 4-7 membered heterocycloalkyl (which 4-7 membered heterocycloalkyl may be substituted with CpC4 alkyl (which CpC4 alkyl may be substituted with aryl, heteroaryl, heterocycloalkyl, CpC4 alkoxy, -O-aryl, -O-heteroaryl, OH), unsubstituted CpC4 haloalkyl, aryl (which aryl may be substituted with halo, unsubstituted CpC4 alkoxy), OH, halo, cyano, acyl (which acyl may be substituted with unsubstituted aryl, unsubstituted heterocycloalkyl, unsubstituted CpC4 alkyl), heteroaryl (which heteroaryl may be substituted with halo), CpC4 dialkylamino, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted -O-heteroaryl, amido (which heteroaryl may be substituted with unsubstituted CpC4 alkyl), unsubstituted CpC4 alkoxy), 5-10 membered heteroaryl (which heteroaryl may be substituted with CpC4 alkyl (which CpC4 alkyl may be substituted with unsubstituted aryl), unsubstituted CpC4 haloalkyl, unsubstituted amido, halo, 4-7-membered heterocycloalkyl (which heterocycloalkyl may be substituted with unsubstituted CpC4 alkyl), unsubstituted CpC4 alkoxy, CN, unsubstituted C3-C7 cycloalkyl, OH, aryl (which aryl may be substituted with unsubstituted CpC4 haloalkyl) unsubstituted 5-7-membered heteroaryl, carboxy (which carboxy may be substituted with unsubstituted CpC4 alkyl)), or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from
Ci-Cβ alkyl (which CpC4 alkyl is substituted with aryl (which aryl may be substituted with halo, CN, unsubstituted CpC4 alkyl, unsubstituted 5-7-membered heterocycloalkyl, unsubstituted 5-10 membered heteroaryl), 5-10 membered heteroaryl (which heteroaryl may be substituted with unsubstituted CpC4 alkyl)), aryl (which aryl may be substituted with halo, CN, unsubstituted C1-C4 alkoxy, unsubstituted amido, unsubstituted CpC4 alkyl, unsubstituted CpC4, haloalkyl, -(CH2) i_4-CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted with ), unsubstituted 4-7 membered heterocycloalkyl, 5-10 membered heteroaryl (which heteroaryl may be substituted with halo, cyano, unsubstituted CpC4 alkyl, unsubstituted CpC4 alkoxy, unsubstituted 4-7 membered heterocycloalkyl, ); each R4a, R4b and R4c is independently selected from H, Ci-Ce alkyl (which CpC4 alkyl may be substituted with unsubstituted CpC4 alkoxy, unsubstituted dialkylamino, unsubstituted 4-7 membered heterocycloalkyl), unsubstituted C3-C7 cycloalkyl, acyl (which acyl may be substituted with unsubstituted Ci-Cβ alkyl); ml is O, I, or 2; m2 is 0, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that when Ll is -O-, -N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1, 2, 3, or 4; or pharmaceutically acceptable salts or solvates thereof, or solvates of the pharmaceutically acceptable salts. ] In a preferred embodiment, the compound is according to Formula I above wherein:
CyI is selected from aryl and heteroaryl; Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)-
, -S(O)2-, - N(R4a)CO-, -CH2-N(R4a)-or - N(R4a)SO2-; each R1 is independently selected from unsubstituted CpC6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted Ci-Ce alkoxy, unsubstituted amido, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from unsubstituted Ci-Cβ alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted CpC6 alkoxy, unsubstituted amido, unsubstituted alkoxycarbonyl, unsubstituted arylalkyloxy, unsubstituted amino, unsubstituted aryl, unsubstituted arylalkyl, aminosulfonyl (which aminosulfonyl may be substituted with unsubstituted CpC4 alkyl), sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, unsubstituted heteroaryl, hydroxyl, nitro, and thiol;
R a is selected from unsubstituted Ci-Cβ alkyl and unsubstituted C3-C7 cycloalkyl;
R3b is independently selected from aryl (which aryl may be substituted with halo, unsubstituted
4-7 membered heterocycloalkyl, unsubstituted CpC4 alkyl, unsubstituted CpC4 alkoxy, unsubstituted CpC4 haloalkyl, unsubstituted CpC4 haloalkoxy, unsubstituted 5-7-membered heteroaryl, unsubstituted acylamino, unsubstituted amino, cyano, -(CH2) I-4-CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted with cyano), 4-7 membered heterocycloalkyl (which 4-7 membered heterocycloalkyl may be substituted with C1-C4 alkyl (which C1-C4 alkyl may be substituted with aryl, heteroaryl, heterocycloalkyl, C1-C4 alkoxy, -O-aryl, -O-heteroaryl, OH), unsubstituted Ci-C4haloalkyl, aryl (which aryl may be substituted with halo, unsubstituted C1-C4 alkoxy), OH, halo, cyano, acyl (which acyl may be substituted with unsubstituted aryl, unsubstituted heterocycloalkyl, unsubstituted CpC4 alkyl), heteroaryl (which heteroaryl may be substituted with halo), CpC4 dialkylamino, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted -O-heteroaryl, amido (which heteroaryl may be substituted with unsubstituted C1-C4 alkyl), unsubstituted C1-C4 alkoxy), 5-10 membered heteroaryl (which heteroaryl may be substituted with C1-C4 alkyl (which C1-C4 alkyl may be substituted with unsubstituted aryl), unsubstituted C1-C4 haloalkyl, unsubstituted amido, halo, 4-7-membered heterocycloalkyl (which heterocycloalkyl may be substituted with unsubstituted CpC4 alkyl), unsubstituted CpC4 alkoxy, CN, unsubstituted C3-C7 cycloalkyl, OH, aryl (which aryl may be substituted with unsubstituted CpC4 haloalkyl) unsubstituted 5-7-membered heteroaryl, carboxy (which carboxy may be substituted with unsubstituted CpC4 alkyl)), or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from Ci-Ce alkyl (which CpC4 alkyl may be substituted with aryl (which aryl may be substituted with halo, CN, unsubstituted CpC4 alkyl, unsubstituted 5-7- membered heterocycloalkyl, unsubstituted 5-10 membered heteroaryl), 5-10 membered heteroaryl (which heteroaryl may be substituted with unsubstituted CpC4 alkyl)), aryl (which aryl may be substituted with halo, CN, unsubstituted CpC4 alkoxy, unsubstituted amido, unsubstituted CpC4 alkyl, unsubstituted CpC4, haloalkyl, -(CH2)M-CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted with ), unsubstituted 4-7 membered heterocycloalkyl, 5-10 membered heteroaryl (which heteroaryl may be substituted with halo, cyano, unsubstituted CpC4 alkyl, unsubstituted CpC4 alkoxy, unsubstituted 4-7 membered heterocycloalkyl, ); each R4a, R4b and R4c is independently selected from H, Ci-Cβ alkyl (which CpC4 alkyl may be substituted with unsubstituted CpC4 alkoxy, unsubstituted dialkylamino, unsubstituted 4-7 membered heterocycloalkyl), unsubstituted C3-C7 cycloalkyl, acyl (which acyl may be substituted with unsubstituted Ci-Ce alkyl); ml is O, 1, or 2; m2 is O, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that when Ll is -O-, -N(R4a)-, -CH2-N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1, 2, 3, or 4; or pharmaceutically acceptable salts or solvates thereof, or solvates of the pharmaceutically acceptable salts. [00130] In one embodiment, with respect to compounds of Formula I, ml is 0.
[00131] In one embodiment, with respect to compounds of Formula I, ml is 1 or 2; each R1 is independently selected from Ci-Cβalkyl, substituted Ci-Cβalkyl, and halo.
[00132] In a particular embodiment, with respect to compounds of Formula I, ml is 1 or 2 and each R1 is independently selected from Me, CF3, Cl and F.
[00133] In one embodiment, with respect to compounds of Formula I, R a is substituted or unsubstituted Ci-Ce alkyl.
[00134] In another embodiment, with respect to compounds of Formula I, R a is substituted or unsubstituted C3-C7 cycloalkyl.
[00135] In a particular embodiment, with respect to compounds of Formula I, R a is cyclopropyl, cyclobutyl, or cyclop entyl.
[00136] In a further embodiment, with respect to compounds of Formula I, R4b and R4c are independently selected from H and Me.
[00137] In a more particular embodiment, with respect to compounds of Formula I, the compound is according to Formula II:
Figure imgf000033_0001
wherein CyI, Ll, R3a, R3b, m2, and nl are as described for Formula I.
[00138] In one embodiment, with respect to compounds of Formula II, CyI is Ph; and m2 is 0.
[00139] In one embodiment, with respect to compounds of Formula II, CyI is Ph; m2 is 1, 2 or
3; and each R a is independently Ci-Cβ alkyl, Ci-Ce haloalkyl, Ci-Cβ alkoxy, or halo.
[00140] In a particular embodiment, with respect to compounds of Formula II, CyI is Ph; m2 is
1, 2 or 3; and each R3a is independently Cl, F, Me, Et, OMe, CF3, CONH2, CONMe2, CONHMe, CN,
NHCOMe, COOH, OH or COOEt.
[00141] In another embodiment, with respect to compounds of Formula II, CyI is 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 benzodioxanyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted quinolinyl, or substituted or unsubstituted isoquinolinyl; and m2 is 0. [00142] In a more particular embodiment, the compound is according to Formula III:
Figure imgf000034_0001
wherein Ll, R3b, and nl are as described for Formula I.
[00143] In one embodiment, with respect to compounds of Formula III, R3b is substituted or unsubstituted aryl, substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted
C3-C7 cycloalkyl, or substituted or unsubstituted 4-7 membered heterocycloalkyl.
[00144] In a particular embodiment, with respect to compounds of Formula III, Ll is selected from a single bond, -O-, -N(R4a)-, -C(O)-, C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -N(R4a)SO2- and -N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted aryl, substituted or unsubstituted 5-10 membered heteroaryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl.
[00145] In another particular embodiment, with respect to compounds of Formula III, Ll is selected from - a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -
N(R4a)SO2- and -N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted C3-C7 cycloalkyl.
[00146] In a more particular embodiment, with respect to compounds of Formula III, Ll is selected from - a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -
SO2N(R4a)-, -N(R4a)SO2- and -N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted cyclopropyl, substituted or unsubstituted cyclobutyl, substituted or unsubstituted cyclohexyl, or substituted or unsubstituted cyclopentyl.
[00147] In one embodiment, with respect to compounds of Formula III, Ll is selected from: a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -N(R4a)SO2- and -
N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted aryl or substituted or unsubstituted 5-10 membered heteroaryl.
[00148] In a particular embodiment, with respect to compounds of Formula III, Ll is selected from - a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -
N(R4a)SO2- and -N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted phenyl, substituted or unsubstituted pyridyl, substituted or unsubstituted pyrrolyl, substituted or unsubstituted pyrazolyl, substituted or unsubstituted imidazolyl, substituted or unsubstituted triazolyl, substituted or unsubstituted tetrazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted oxadiazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted thiophenyl, substituted or unsubstituted indolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted benzofuranyl, substituted or unsubstituted benzodioxanyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted quinolinyl, or substituted or unsubstituted isoquinolinyl.
[00149] In one embodiment, with respect to compounds of Formula III, Ll is selected from: a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -N(R4a)SO2- and -
N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is substituted or unsubstituted 4-7-membered heterocycloalkyl, provided that when the heterocycle is attached via a heteroatom, and Ll is -O-, -N(R4a)-, -SO2N(R4a)-, and -CON(R4a)-, nl is not 0 or 1.
[00150] In a particular embodiment, with respect to compounds of Frmula III, Ll is selected from: a single bond, -O-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -CON(R4a)-, -SO2N(R4a)-, -S(O)2-, -N(R4a)SO2- and -N(R4a)CO-; nl is 0, 1, 2, 3, or 4; and R3b is piperidinyl, morpholinyl, piperazinyl, homopiperazinyl or pyrrolidinyl, each of which may be unsubstituted or substituted with CpC6 alkyl, acyl, phenyl, or OH, provided that when the heterocycle is attached via an heteroatom, and Ll is -O-, -N(R4a)-, -SO2N(R4a)-, and -CON(R4a)-, nl is not 0 or 1.
[00151] In one particular embodiment, with respect to compounds of Formula III, Ll is a single bond.
[00152] In another particular embodiment, with respect to compounds of Formula III, Ll is selected from -O-, and -N(R4a)-.
[00153] In another particular embodiment, with respect to compounds of Formula III, Ll is selected from -C(O)-, and -S(O)2-.
[00154] In another particular embodiment, with respect to compounds of Formula III, Ll is selected from -CON(R4a)-, and -SO2N(R4a)-.
[00155] In another particular embodiment, with respect to compounds of Formula III, Ll is selected from -N(R4a)SO2- and -N(R4a)CO-.
[00156] In one particular embodiment, with respect to compounds of Formula III, Ll is -
C[=N(R4a)]-,
[00157] In a further aspect of the invention R4a is H, substituted or unsubstituted CpC4 alkyl, substituted or unsubstituted CpC4 alkoxy.
[00158] In one embodiment, with respect to compounds of Formula I, R4a is H
[00159] In one embodiment, with respect to compounds of Formula I, R4a is -(CH2)n2-R6a; wherein n2 is 0, 1, 2 and R6a is H, CN, NMe2, or tetrahydrofuranyl.
[00160] In another embodiment, with respect to compounds of Formula I, R4a is -CH(CH3)-
(CH2)n2-R6a; wherein n2 is 0 or 1 and R6a is H, or OMe.
[00161] In another embodiment, with respect to compounds of Formula I, R3b is OPh,and O-(4-
F-Ph). [00162] In another embodiment, with respect to compounds of Formula I, R3b is CO-R3c; and R3c is:
Figure imgf000036_0001
[00163] In one embodiment, the compound is according to formula III, and -Ph-Ll -(CH2)ni-R3 is:
Figure imgf000036_0002
wherein n2 is nl; and R3b, and nl are as described for Formula 1 ; and Cy3 is a substituted or unsubstituted nitrogen containing 4-7-membered heterocycloalkyl group.
[00164] In another embodiment, the compound is according to Formula III, and -Ph-Ll -(CH2)ni-
R3b is:
Figure imgf000036_0003
wherein n2 is nl; and R , 3b , and nl are as described for Formula 1; and Cy3 is a substituted or unsubstituted nitrogen containing 4-7-membered heterocycloalkyl group.
[00165] In one embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and R3b is unsubstituted aryl. [00166] In another embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and R3b is substituted aryl.
[00167] In another embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and R3b is
Figure imgf000037_0001
and one of R49 and R50 may be hydrogen and at least one of R49 and R50 is each independently selected from CpCg alkyl, 4-10 membered heterocycloalkyl, CpCg alkoxy, hetero-O-aryl, alkylamino,
NR51COR52, NR51SOR52 NR51SO2R52, COOalkyl, COOaryl, CONR51R52, CONR51OR52, NR51R52,
SO2NR51R52, S-alkyl, SOalkyl, S02alkyl, Saryl, SOaryl, S02aryl; or R49 and R50 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. R51, and R52 are independently hydrogen, Ci-Cg alkyl, CpC4 haloalkyl, C3-Ci0 cycloalkyl, 4-10 membered heterocycloalkyl, C6-Ci0 aryl, substituted aryl, 5-10 membered heteroaryl.
[00168] In another embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and R3b is aryl, substituted with one or more of groups selected from halo, CpCg alkyl, Cp
C8 haloalkyl, Ci-Cg haloalkoxy, cyano, hydroxy, Ci-Cg alkoxy, and amino.
[00169] In another embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and R3b is phenyl, substituted with one or more of groups selected from halo, Ci-Cg alkyl,
CpCg haloalkyl, CpCg haloalkoxy, cyano, hydroxy, CpCg alkoxy, and amino.
[00170] In one embodiment, the-Ph-Ll-(CH2)npR3b and R3b are as described in the preceding paragraph, and the substitution is other than 3-OMe.
[00171] In another embodiment, the compound is other than N-[5-[4-[(3-methoxyphenyl)- methoxy]phenyl][l,2,4]triazolo[l,5-a]pyridin-2-yl]-cyclopropanecarboxamide.
[00172] In one particular embodiment, the -Ph-Ll -(CH2)npR3b group is as described in the preceding paragraphs, and R3b is substituted or unsubstituted heteroaryl.
[00173] In another embodiment, the -Ph-Ll -(CH2)npR3b is as described in the preceding paragraphs, and R3b is unsubstituted thiophenyl, pyrrolyl, benzothiophenyl, benzofuranyl, indolyl, pyridyl, quinolinyl, imidazolyl, oxazolyl and pyrazinyl
[00174] In another embodiment, the -Ph-Ll -(CH2)npR3b group is as described in the preceding paragraphs, and R3b is thiophenyl, pyrrolyl, benzothiophenyl, benzofuranyl, indolyl, pyridyl, quinolinyl, imidazolyl, oxazoleyl and pyrazineyl, substituted with one or more groups selected from halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR"'SO2R", -SO2NR R", -C(O)R", -C(O)OR", -
OC(O)R", -NR"'C(O)R", -C(O)NR"R ", -NR11R1", -(CR"'R"')mOR"', wherein, each R" is independently selected from H, C1-C8 alkyl, -(CH2MC6-C10 aryl), -(CH2)t(5-10 membered heteroaryl), -(CH2X(C3-Ci0 cycloalkyl), and -(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4.
[00175] In another embodiment, the -Ph-Ll -(CH2)nrR3b group is as described in the preceding paragraphs, and R3b is unsubstituted pyridyl.
[00176] In another embodiment, the -Ph-Ll -(CH2)nrR3b group is as described in the preceding paragraphs, and R3b is pyridyl, substituted with one or more groups selected from halo, cyano, nitro, trifluoromethyl, trifluoromethoxy, azido, -NR"'SO2R", -SO2NR R", -C(O)R", -C(O)OR", -OC(O)R", -
NR'"C(O)R", -C(O)NR R", -NR"R ", -(CR'"R'")mOR'", wherein, each R" is independently selected from
H, Ci-C8 alkyl, -(CH2MC6-C10 aryl), -(CH2)t(5-10 membered heteroaryl), -(CH2MC3-C10 cycloalkyl), and
-(CH2)t(4-10 membered heterocycloalkyl), wherein t is an integer from 0 to 4.
[00177] In another embodiment, the -Ph-Ll -(CH2)nrR3b is as described in the preceding paragraphs, and R3b is pyridyl, substituted with halo, cyano, methyl, or trifluoromethyl.
[00178] In another embodiment, the -Ph-Ll -(CH2)ni-R3b is as described in the preceding paragraphs, and nl or n2 is 1.
[00179] In another embodiment, the -Ph-Ll -(CH2)nrR3b is as described in the preceding paragraphs, and nl or n2 is 2.
[00180] In a particular embodiment, the compound is according to Formula III and R3b is selected from:
Figure imgf000038_0001
wherein each R5a is independently C1-C4 alkyl, halo, CF3 or Phenyl; R5b is H, aryl, 5-10 membered heteroaryl, heteroaryl, C3-C6 cycloalkyl, or 4-7 membered heterocycloalkyl; and m5 is 0, 1 or 2. [00181] In another particular embodiment, the compound is according to Formula III and R3b is selected from:
Figure imgf000039_0001
wherein each R5a is independently C1-C4 alkyl, halo, oxo, CF3 or Phenyl; R5b is H, C1-C4 alkyl, aryl, 5-10 membered heteroaryl, heteroaryl, C3-C6 cycloalkyl, or 4-7 membered heterocycloalkyl; and m5 is 0, 1 or
2.
[00182] In another particular embodiment, the compound is according to Formula III and Cy3 is selected from:
Figure imgf000039_0002
wherein each R5a is independently C1-C4 alkyl, halo, CF3 or Phenyl; R5b is H, aryl, 5-10 membered heteroaryl heteroaryl, C3-C6 cycloalkyl, or 4-7 membered heterocycloalkyl; R5c is H, or CpC4 alkyl; m5 is O, 1, or 2; n5 is O, I, or 2.
[00183] In a more particular embodiment, the compound is according to any one of Formulae
IVa, IVb, IVc, IVd, IVe, or IVf:
Figure imgf000040_0001
[00184] In a more particular embodiment, the compound is according to any one of Formulae
Va, Vb, Vc, or Vd:
Figure imgf000040_0002
[00185] In a more particular embodiment, the compound is according to any one of Formulae
Via, VIb, VIc, or VId:
Figure imgf000041_0001
[00186] In a more particular embodiment, the compound is according to any one of Formulae
Vila, VIIb, VIIc, VIId, VIIe, or VIIf:
Figure imgf000041_0002
Figure imgf000041_0003
[00187] In another more particular embodiment, the compound is according to any one of
Formulae Villa, VIIIb, VIIIc, VIIId, VIIIe, VIIIf, VIIIg, VIIIh, Villi, VIIIj, VIIIk or Villi:
Figure imgf000042_0001
[00188] In another more particular embodiment, the compound is according to any one of
Formulae IXa, IXb, IXc, IXd, IXe, IXf, IXg, IXh, IXi, IXj, IXk or IXl:
Figure imgf000043_0001
[00189] In certain aspects, the present invention provides compounds according to Formula X, or XI:
Figure imgf000043_0002
wherein R3b is substituted or unsubstituted 4-7 membered heterocycloalkyl; provided that when the compound is according to Formula X, the heterocycloalkyl ring is other than unsubstituted morpholin-1- yi.
[00190] In one embodiment, the compound is according to Formula X. In another embodiment, the compound is according to Formula XL
[00191] In one particular embodiment, with respect to compounds according to Formula X, R3b is unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, azepinyl, pyrrolidonyl, pyranyl, dihydrothiophenyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothiophenyl, dioxanyl, tetrahydropyranyl, imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 2-pyrazolineyl, pyrazolidinyl, thiomorpholinyl-S-oxide, and thiomorpholinyl-S,S-dioxide piperidonyl, or piperazonyl.
[00192] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, thiomorpholin-l-yl-S,S-dioxide, piperazin-1- yl, or azepin-1-yl.
[00193] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted azetidin-1-yl.
[00194] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted pyrrolidin-1-yl.
[00195] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted piperidin-1-yl or piperazin-1-yl.
[00196] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted thiomorpholin-1 -yl-S,S-dioxide.
[00197] In one particular embodiment, with respect to compounds of Formula X, R3b is unsubstituted azepin-1-yl.
[00198] In one particular embodiment, with respect to compounds of Formula X, R3b is azetidin-
1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1 -yl, piperazin-1-yl, or azepin-1-yl; substituted with one or more group s elected from C1-C4 alkyl, C1-C4 haloalkyl, cyano, amino, dialkylamino, dialkylaminomethyl, hydroxy, halo, acyl, acylamino, C1-C4 hydroxyalkyl, C1-C4 alkoxy, carboxamido, and C1-C4 dialkyl carboxamido.
[00199] In one particular embodiment, with respect to compounds of Formula X, R3b is azetidin-
1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1 -yl, piperazin-1-yl, or azepin-1-yl; substituted with Me, CF3, F, Cl, difluoro, dimethyl, hydroxy, cyano, dimethylamino, dimethylaminomethyl, hydroxymethyl, carboxamido, N,N-dimethylcarboxamido, methoxy, ethoxy, or 2,2,2,-trifluoroethyl. [00200] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, azepinyl, pyrrolidonyl, pyranyl, dihydrothiophenyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothiophenyl, dioxanyl, tetrahydropyranyl, imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 2-pyrazolineyl, pyrazolidinyl, morpholinyl, thiomorpholinyl-S-oxide, and thiomorpholinyl-
S,S-dioxide piperidonyl, or piperazonyl.
[00201] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholinyl, thiomorpholin-l-yl-S,S- dioxide, piperazin-1-yl, or azepin-1-yl.
[00202] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted azetidin-1-yl.
[00203] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted pyrrolidin-1-yl.
[00204] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted piperidin-1-yl or piperazin-1-yl.
[00205] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted thiomorpholin-1 -yl-S,S-dioxide.
[00206] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted morpholin-1-yl.
[00207] In one particular embodiment, with respect to compounds of Formula XI, R3b is unsubstituted azepin-1-yl.
[00208] In one particular embodiment, with respect to compounds of Formula XI, R3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl, piperazin-1-yl, or azepin-1-yl; substituted with one or more group selected from C1-C4 alkyl, C1-C4 haloalkyl, cyano, amino, dialkylamino, dialkylaminomethyl, hydroxy, halo, acyl, acylamino, C1-C4 hydroxyalkyl, C1-C4 alkoxy, carboxamido, and C1-C4 dialkyl carboxamido.
[00209] In one particular embodiment, with respect to compounds of Formula XI, R3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1-yl, piperazin-1-yl, or azepin-1-yl; substituted with Me, CF3, F, Cl, difluoro, dimethyl, hydroxy, cyano, dimethylamino, dimethylaminomethyl, hydroxymethyl, carboxamido, N,N-dimethylcarboxamido, methoxy, ethoxy, or 2,2,2,-trifluoroethyl.
[00210] In one embodiment, with respect to Formula I, the compound is selected from the compounds exemplified in Table 1.
[00211] In one embodiment the compound of the invention is not an isotopic variant.
[00212] In one aspect a compound of the invention according to any one of the embodiments herein described is present as the free base.
[00213] In one aspect a compound of the invention according to any one of the embodiments herein described is a pharmaceutically acceptable salt.
[00214] In one aspect a compound of the invention according to any one of the embodiments herein described is a solvate of the compound. [00215] In one aspect a compound of the invention according to any one of the embodiments herein described is a solvate of a pharmaceutically acceptable salt of the compound. [00216] While specified groups for each embodiment have generally been listed above separately, a compound of the invention includes one in which several or each embodiment in the above Formula, as well as other formulae presented herein, is selected from one or more of particular members or groups designated respectively, for each variable. Therefore, this invention is intended to include all combinations of such embodiments within its scope.
[00217] In certain aspects, the present invention additionally 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.
[00218] Other derivatives of the compounds of the invention have activity in both their acid and acid derivative forms, but the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985). 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, C2- Cg alkenyl, aryl, C7-C12 substituted aryl, and C7-C12 arylalkyl esters of the compounds of the invention.
PHARMACEUTICAL COMPOSITIONS
[00219] When employed as pharmaceuticals, the compounds of the 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. Generally, 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.
[00220] The pharmaceutical compositions of the invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal. Depending on the intended route of delivery, 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
[00221] 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. The term "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 prefϊlled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such 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.
[00222] 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. [00223] Injectable compositions are typically based upon injectable sterile saline or phosphate- buffered saline or other injectable carriers known in the art. As before, 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.
[00224] 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. 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. [00225] The compounds of this invention can also be administered by a transdermal device.
Accordingly, transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
[00226] The above-described components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack
Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
[00227] The compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.
[00228] The following formulation examples illustrate representative pharmaceutical compositions that may be prepared in accordance with this invention. The present invention, however, is not limited to the following pharmaceutical compositions.
Formulation 1 - Tablets
[00229] 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
[00230] 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).
Formulation 3 - Liquid
[00231] 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.
Formulation 4 - Tablets
[00232] 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.
Formulation 5 - Injection
[00233] 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. Formulation 6 - Topical
[00234] 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) may be added and the resulting mixture is stirred until it congeals.
METHODS OF TREATMENT
[00235] The present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to aberrant activity of JAK. In particular, conditions related to aberrant activity of one or more of JAKl, JAK2, JAK3 and/or TYK2. Accordingly, the 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. 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection). Inhibitors of JAK can also find application in the treatment of proliferative diseases. In particular the inhibitors of JAK find application in the treatment of cancers, especially leukaemias and solid tumours (e.g. uterine leiomyosarcoma, prostate cancer). In particular the conditions are selected from inflammatory conditions, conditions related to cartilage and/or joint degradation in mammals including humans. In another embodiment, the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating proliferative disorders in mammals, including humans. In a specific embodiment the compound of the invention and pharmaceutical compositions thereof find use as therapeutics for preventing and/or treating cancer in mammals including humans. [00236] In additional method of treatment aspects, 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 a compound of the invention herein described. In a specific embodiment, the autoimmune disease is selected from COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and inflammatory bowel disease.
[00237] In another aspect 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. In a specific embodiment, the autoimmune disease is selected from COPD, asthma, systemic lupus erythematosis, type I diabetes mellitus and inflammatory bowel disease. [00238] In a method of treatment aspect, 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 therapeutically effective amount of a compound of the invention, or one or more of the pharmaceutical compositions or compounds herein described.
[00239] In another aspect 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.
[00240] 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 an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.
[00241] In another aspect 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.
[00242] In another aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition involving inflammation, which method comprises administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described. In additional method of treatment aspects, 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. 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, which methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.. In a specific embodiment, 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.
[00243] In another aspect, this invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of a condition involving inflammation. In another aspect 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. In a specific embodiment, the condition involving inflammation is selected from rheumatoid arthritis, osteoarthritis, allergic airway disease (e.g. asthma) and inflammatory bowel diseases.
[00244] In further method of treatment aspects, 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 methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described. In further method of treatment aspects, 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 methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.
[00245] In another aspect 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. In another aspect 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. [00246] In further method of treatment aspects, 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 methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described..
[00247] In another aspect 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
Castleman's disease or mesangial proliferative glomerulonephritis.
[00248] In further method of treatment aspects, this invention provides methods of treating a mammal susceptible to or afflicted with transplantation rejection, which methods comprise administering an effective amount of one or more of the compounds of the invention or the pharmaceutical compositions herein described.. In a specific embodiment, the invention provides methods of treating organ transplant rejection.
[00249] In another aspect the present invention provides a compound of the invention for use in the treatment, prevention or prophylaxis of transplantation rejection. In a specific embodiment, the invention provides methods of treating organ transplant rejection.
[00250] As a further aspect of the invention there is provided 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.
[00251] 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 particular 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.
[00252] 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 particular 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. In a particular embodiment said compounds exhibit cartilage anabolic and/or anti-catabolic properties. [00253] 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. [00254] For the prevention and/or treatment of long-term conditions, such as degenerative conditions, the regimen for treatment usually stretches over many months or years so oral dosing is preferred for patient convenience and tolerance. With oral dosing, one to five and especially two to four and typically three oral doses per day are representative regimens. Using these dosing patterns, 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. [00255] Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
[00256] When used to prevent the onset of an inflammatory condition, the compounds of this 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.
[00257] 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 be 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. [00258] In one embodiment, a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of a disease involving inflammation; particular 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.
[00259] In one embodiment, 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 cyclosporin), and biological DMARDS (for example but without limitation Infliximab, Etanercept, Adalimumab, Rituximab, and Abatacept).
[00260] In one embodiment, 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. Herceptin™), capecitabine, raloxifene hydrochloride, EGFR inhibitors (e.g. lressa®, Tarceva™, Erbitux™), VEGF inhibitors (e.g. Avastin™), proteasome inhibitors (e.g. Velcade™), Glivec® or hsp90 inhibitors (e.g. 17-AAG). Additionally, a compound of the invention may be administered in combination with other therapies including, but not limited to, radiotherapy or surgery. In a specific embodiment the proliferative disorder is selected from cancer, myeloproliferative disease or leukaemia. [00261] In one embodiment, a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of autoimmune diseases, particular 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-β), TNF binding proteins (e.g. infliximab (Remicade), etanercept (Enbrel), or adalimumab (Humira)), mycophenolate, Fingolimod, Myriocin. [00262] In one embodiment, a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of transplantation rejection, particular 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-2Rα receptor antibodies, basiliximab, daclizumab), polyclonal anti-T-cell antibodies (e.g. anti-thymocyte globulin (ATG), anti-lymphocyte globulin (ALG)).
[00263] In one embodiment, 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: beta2-adrenoceptor agonists (e.g. salbutamol, levalbuterol, terbutaline and bitolterol.), epinephrine (inhaled or tablets), anticholinergics (e.g. ipratropium bromide), glucocorticoids (oral or inhaled) Long-acting β2-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). [00264] Additionally, 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. 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.
[00265] In one embodiment, a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of IBD, particular 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 cyclosporin) and biological disease modifying, immunomodulatory agents (infliximab, adalimumab, rituximab, and abatacept).
[00266] In one embodiment, 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.
[00267] In one embodiment, a compound of the invention is co-administered with another therapeutic agent for the treatment and/or prevention of psoriasis, particular 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 D3 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, Raptiva and ustekinumab (a IL- 12 and IL-23 blocker). Additionally, a compound of the invention may be administered in combination with other therapies including, but not limited to phototherapy, or photochemotherapy (e.g. psoralen and ultraviolet A phototherapy (PUVA)). [00268] By co-administration 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.
GENERAL SYNTHETIC PROCEDURES General
[00269] 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.
[00270] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein. [00271] The following methods are presented with details as to the preparation of representative bicycloheteroaryls that have been listed hereinabove. The compounds of the invention may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.
[00272] All reagents were of commercial grade and were used as received without further purification, unless otherwise stated. Commercially available anhydrous solvents were used for reactions conducted under inert atmosphere. Reagent grade solvents were used in all other cases, unless otherwise specified. Column chromatography was performed on silica gel 60 (35-70 μm). Thin layer chromatography was carried out using pre-coated silica gel F-254 plates (thickness 0.25 mm). 1H NMR spectra were recorded on a Bruker DPX 400 NMR spectrometer (400 MHz). Chemical shifts (δ) for 1H NMR spectra are reported in parts per million (ppm) relative to tetramethylsilane (δ 0.00) or the appropriate residual solvent peak, i.e. CHCI3 (δ 7.27), as internal reference. Multiplicities are given as singlet (s), doublet (d), triplet (t), quartet (q), multiplet (m) and broad (br). Coupling constants (J) are given in Hz. Electrospray MS spectra were obtained on a Micromass platform LC/MS spectrometer. Column Used for all LCMS analysis: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350)). Preparative HPLC :Waters XBridge Prep C18 5μm ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
[00273] List of abbreviations used in the experimental section:
Figure imgf000057_0002
Figure imgf000057_0001
Synthetic Preparation of Compounds of the Invention
[00274] A compound of the invention can be produced according to the following scheme.
Preparation of the core intermediate Scheme 1
Figure imgf000058_0001
wherein Ar is Cyl-Ll-(CR4bR4c)nl-R3b; and CyI, Ll, nl, R2a, R3b, R4b, and R4c are as described herein.
1.1.1 l-(6-Bromo-pyridin-2-yl)-3-carboethoxy-thiourea (2)
Figure imgf000058_0002
[00275] To a solution of 2-amino-6-bromopyridine (1) (253.8 g, 1.467 mol) in DCM (2.5 L) cooled to 5 0C was added ethoxycarbonyl isothiocyanate (173.0 mL, 1.467 mol) dropwise over 15 min. The reaction mixture was then allowed to warm to room temp. (20 0C) and stirred for 16 h. Evaporation in vacuo gave a solid which was collected by filtration, thoroughly washed with petrol (3 x600 mL) and air-dried to afford (2). The thiourea was used as such for the next step without any purification. 1H (400 MHz, CDCl3) δ 12.03 (IH, br s, NH), 8.81 (IH, d, J 7.8 Hz, H-3), 8.15 (IH, br s, NH), 7.60 (IH, t, J 8.0 Hz, H-4), 7.32 (IH, dd, J 1.1 and 0.6 Hz, H-5), 4.31 (2H, q, J 7.1 Hz, CH2), 1.35 (3H, t, J 7.1 Hz, CH3).
7.7.2 5-Bromo-[l,2,4]triazolo[l,5-a]pyridin-2-ylamine (3)
Figure imgf000058_0003
[00276] To a suspension of hydroxylamine hydrochloride (101.8 g, 1.465 mol) in EtOH/MeOH
(1 :1, 900 mL) was added N,N-diisopropylethylamine (145.3 mL, 0.879 mol) and the mixture was stirred at room temp. (20 0C) for 1 h. l-(6-Bromo-pyridin-2-yl)-3-carboethoxy-thiourea (2) (89.0 g, 0.293 mol) was then added and the mixture slowly heated to reflux (Note: bleach scrubber is required to quench H2S evolved). After 3 h at reflux, the mixture was allowed to cool and filtered to collect the precipitated solid. Further product were collected by evaporation in vacuo of the filtrate, addition of H2O (250 mL) and filtration. The combined solids were washed successively with H2O (250 mL), EtOH/MeOH (1 :1, 250 mL) and Et2O (250 mL) then dried in vacuo to afford the triazolopyridine derivative (3) as a solid. The compound was used as such for the next step without any purification. 1H (400 MHz,
Figure imgf000059_0001
δ 7.43-7.34 (2H, m, 2 x aromatic-H), 7.24 (IH, dd, J 6.8 and 1.8 Hz, aromatic-H), 6.30 (2H, br, NH2); m/z 213/215 (1 :1, M+H+, 100%).
1.1.3 Procedure for mono-acylation to afford intermediateCyclopropanecarboxylic acid (5- bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide
Figure imgf000059_0002
[00277] To a solution of the 2-amino-triazolopyridine (3) (7.10 g, 33.3 mmol) in dry CH3CN
(150 mL) at 5 0C was added Et3N (11.6 mL, 83.3 mmol) followed by cyclopropanecarbonyl chloride (83.3 mmol). The reaction mixture was then allowed to warm to ambient temperature and stirred until all starting material (3) was consumed. If required, further Et3N (4.64 mL, 33.3 mmol) and the acid chloride (33.3 mmol) were added to ensure complete reaction. Following solvent evaporation in vacuo the resultant residue was treated with 7 N methanolic ammonia solution (50 mL) and stirred at ambient temp, for 1 h to hydro lyse any bis-acylated product. Product isolation was made by removal of volatiles in vacuo followed by trituration with Et2O (50 mL). The solids were collected by filtration, washed with H2O (2χ50mL), acetone (50 mL) and Et2O (50 mL), then dried in vacuo to give the required acyl intermediate (4).
Method A
1.1.4 Preparation of compounds of the invention via Suzuki coupling (5):
Figure imgf000059_0003
[00278] An appropriate boronic acid (2eq.) is added to a solution of bromo intermediate in 1,4- dioxane/water (5:1). K2CO3 (2 eq.) and PdCl2dppf (5%) are added to the solution. The resulting mixture is then heated in a microwave at 140 0C for 30 min (This reaction can also be carried out by traditional heating in an oil bath at 900C for 16h under N2). Water is added and the solution is extracted with ethyl acetate. The organic layers are dried over MgSθ4 and evaporated in vacuo. The final compound is obtained after purification by flash chromatography. Method B
Figure imgf000060_0001
wherein R4a, R4b, R4c, R3b and nl are as described herein.
Bl. 4 4-[2-(Cyclopropanecarbonyl-amino)-[l ,2,4]triazolo[l ,5-a]pyridin-5-yl] -benzoyl chloride
Figure imgf000060_0002
[00279] 2 Drops of DMF were added to a solution of 4-[2-(cyclopropanecarbonyl-amino)-
[l,2,4]triazolo[l,5-a]pyridin-5-yl]-benzoic acid (1 eq) obtained by Method A in DCM under N2 atmosphere. Then oxalyl chloride (2 eq) was added drop wise to this resulting solution (gas release). The mixture was stirred at room temperature for 2 hours. After completion of the reaction by LCMS, the solvent was removed. The crude acid chloride was used without further purification in next step.
B2. Amide formation (General Method)
Figure imgf000060_0003
[00280] An appropriate amine (1.1 eq; R2b, R2c and ml are as described herein) and Et3N (5 eq) are dissolved in DCM under N2 atmosphere and cooled at 00C. The acid chloride (Bl, 1 eq) dissolved in DCM is added dropwise to this solution. The reaction is stirred at room temperature for 3h. After this time, reaction is complete. The compound is extracted with EtOAc and water, washed with brine and dried over MgSO^ Organic layers are filtered and evaporated. The final compound is isolated by preparative HPLC. Preparative HPLC: Waters XBridge Prep C18 5μm ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method C
3b
Figure imgf000061_0001
wherein R4a,R4b, R4c,R3b and nl are as described herein.
Reaction of alkylation (General Method)
Figure imgf000061_0002
[00281] Cyclopropanecarboxylic acid [5-(4-hydroxy-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]- amide (1.1 eq) obtained by Method A and K2CO3 (5 eq) (or AgCO3) are dissolved in DMF under N2 and the appropriate alkylating agent (1.1 eq) is added dropwise. The resulting suspension is heated at 500C for 16h. After this time, the reaction is complete. The compound is extracted with EtOAc and water, washed with brine and dried over MgSO4. Organic layers are filtered and evaporated. The final compound is isolated by preparative HPLC. Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method D
Figure imgf000062_0001
Coupling reaction (general method)
Figure imgf000062_0002
wherein R4a,R4b, R4c,R3b and nl are as described herein and L is -C(=O)- or -SO2-.
[00282] The aniline derivative (1 eq.) obtained by Method A and Et3N (5 eq) are dissolved in
DCM under N2 and cooled at 00C. The appropriate acid chloride (for A) or sulfonyl chloride (for B) (1.5 eq.) dissolved in DCM is added dropwise to this solution. The reaction is stirred at room temperature for 16 h. After this time, the reaction is complete. The compound is extracted with EtOAc and water, washed with brine and dried over MgSO4. Organic layers are filtered and evaporated. The final compound is isolated by preparative HPLC. Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method E
Figure imgf000062_0003
wherein R4a,R4b, R4c,R3b and nl are as described herein.
Reductive animation (General Method)
Figure imgf000063_0001
[00283] The appropriate aldehyde (2 eq.), the aniline derivative (1 eq.) obtained by Method A and Ti(OPr)4 are mixed and stirred at room temperature for 3 hrs. The mixture is diluted in ethanol and Na(CN)BH3 (leq.) was added. The resulting solution is stirred at room temperature for 16 hrs. The mixture is diluted in water and filtered. The filtrate is washed with ethanol. The combined solvent phases are evaporated under vacuum. The final compound is isolated by preparative HPLC. [00284] Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part
No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method F
Figure imgf000063_0002
wherein Ar is Cyl-Ll-(CR4bR4c)nl-R3b; and CyI, Ll, nl, R3b, R4b, and R4c are as described herein.
N-(5-Bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-acetamide
Figure imgf000063_0003
[00285] To a solution of the 5-bromo-2-amino-triazolopyridine (1 eq.) in dry CH3CN at 5 0C is added Et3N (2.5 eq.) followed by acetyl chloride (2.5 eq.). The reaction mixture is then allowed to warm to ambient temperature and stirred until all starting material is consumed. If required, further Et3N (1 eq.) and acid chloride (1 eq.) are added to ensure complete reaction. Following solvent evaporation in vacuo the resultant residue is treated with 7 N methanolic ammonia solution and stirred at ambient temp, (for 16 h) to hydro lyse any bis-acylated product. Product isolation is made by removal of volatiles in vacuo followed by addition of water and extraction with ethyl acetate. The organic phase is then dried over MgSO4, evaporated in vacuo. The compound may be used without further purification.
Suzuki reaction (General Method)
Figure imgf000064_0001
[00286] The boronic acid (2eq.) is added to a solution of N-(5-Bromo-[l,2,4]triazolo[l,5- a]pyridin-2-yl)-acetamide in 1 ,4-Dioxane/water (5:1). K2CO3 (2 eq.) and Pd(dppf)Cl2 (5%) (dppf = 1,1 '- Bis(diphenylphosphino)ferrocene) are added to the solution. The resulting mixture is then heated in a microwave oven (CEM discover) in a sealed tube at 140 0C for 30 min. Water is added and the solution is extracted with ethyl acetate. The organic layers are dried over MgSθ4 and evaporated in vacuo. The final compound is obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH C18 1.7μm, 2.1mm ID x 50mm L (Part No.186002350).
[00287] Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part
No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method G
Figure imgf000064_0002
Suzuki reaction (general method)
[00288] The appropriate boronic acid (2eq.) is added to a solution of cyclopropanecarboxylic acid [5-(6-chloro-pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide obtained by Method A (1 eq.) i n 1 , 4-dioxane/water (5:1). K2CO3 (2 eq.) and Pd(dppf)Cl2 (5%) (dppf = 1,1 '- Bis(diphenylphosphino)ferrocene) are added to the solution. The resulting mixture is then heated in a microwave oven (CEM discover) in a sealed tube at 140 0C for 30 min. Water is added and the solution is extracted with ethyl acetate. The organic layers are dried over MgSθ4 and evaporated in vacuo. The final compound is obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH C18 1.7μm, 2.1mm ID x 50mm L (Part No.186002350)
[00289] Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part
No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method H
Figure imgf000065_0001
wherein R ,R , R ,R and nl are as described herein.
Aromatic nucleophilic substitution (General Method)
[00290] The chloropyridine derivative obtained by Method A (1 eq), an appropriate amine (1.5 eq.) are mixed in fert-butanol in a sealed tube. The reaction is heated at 90 0C for 24 hours. Once all the SM disappeared by LCMS, water is added to the reaction mixture and the organics is extracted with ethyl acetate. The organic layer is dried over MgSθ4 and evaporater under vacuum. The final compound is isolated by preparative HPLC. Analytical: Waters Acquity UPLC BEH C18 1.7μm, 2.1mm ID x 50mm L (Part No.186002350)
[00291] Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part
No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method I
Figure imgf000065_0002
Reductive alkylation (general method)
[00292] An appropriate amine (2 eq.), cyclopropanecarboxylic acid (for example cyclopropanecarboxylic acid [5-(4-formyl-phenyl)-[l,2,4]triazolo[l,5-a] Dyridine-2-yl]-amide) prepared by method A (1 eq.) and Ti(OPr)4 are mixed and stirred at room temperature for 3 hrs. The mixture is diluted in ethanol and Na(CN)BH3 (leq.) is added. The resulting solution is stirred at room temperature for 16 hrs. The mixture is diluted in water and filtered. The solid is washed with ethanol. The combined solvent phases are evaporated under vacuum. The final compound is isolated by preparative HPLC.
Method J
Figure imgf000066_0001
Reaction of alkylation
Figure imgf000066_0002
[00293] 2-(4-Bromomethyl-phenyl)-4,4,5,5-tetramethyl-[l,3,2]dioxaborolane (leq) and Et3N (2 eq) (or AgCO3) are dissolved in DCM/MeOH (4:1 v:v) under N2 and an amine (2 eq) is added dropwise. The resulting solution is stirred at room temperature for 16h. After this time, the reaction is complete. The solvent is evaporated. The compound is extracted with EtOAc and water, washed with brine and dried over MgSO4. Organic layers are filtered and evaporated. The final compound is isolated by flash chromatography.
Suzuki coupling
Figure imgf000066_0003
The title compound is then synthesized using method A.
Method K
Figure imgf000067_0001
K.I Cyclopropanecarboxylic acid (5-trimethylsilanylethynyl-[ 1,2,4] triazolo[l,5-a] pyridin-
2-yl)-amide
Figure imgf000067_0002
[00294] To a degassed solution of cyclopropanecarboxylic acid (5-bromo-[l,2,4]triazolo[l,5- a]pyridin-2-yl)-amide (0.36 mmol) in THF (3.5 mL) are added CuI (0.036 mmol), Pd(PPh3)2Cl2 (0.036 mmol), (iPr)2NH (0.137 mL), and timethylsilylacetylene (0.43 mmol). The reaction is heated at reflux overnight (700C), and then the solvent is removed under vacuum. The crude is redissolved with ethyl acetate and washed with water. The organic layer is dried over MgSθ4, filtered and the solvent is removed under vacuum to afford the title compound (95 mg, 89% yield). No further purification is carried out.
K.2 Cyclopropanecarboxylic acid (5-ethynyl-[l ,2,4]triazolo[l ,5-a]pyridin-2-yl)-amide
Figure imgf000067_0003
[00295] TBAF (0.4 mmol) IM solution in THF is added to a solution of cyclopropanecarboxylic acid (5-trimethylsilanylethynyl-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide (0.32 mmol) in acetonitrile (4 mL) at room temperature. The reaction mixture is stirred at room temperature until all the starting material disappears by LCMS. The solvent of the reaction is removed under vacuum, and the mixture is redissolved in ethyl acetate. The organic phase is washed with water. The organic layer is dried over MgSO/i, filtered and the solvent is removed under vacuum to afford the pure product (70 mg, 97% yield). No further purification is carried out on the product.
K.3 Cycloaddition (general method)
Figure imgf000068_0001
[00296] The corresponding azide derivative (0.44 mmol) is added to a solution of cyclopropanecarboxylic acid (5-ethynyl-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide (0.44 mmol), CuSO4.5H2O (0.022 mmol) and sodium ascorbate (0.044 mmol) in CHCl3/EtOH/H2O (9:1 :1) at room temperature. The reaction mixture is heated at 500C until completion of the reaction (monitored by LCMS). The crude mixture is diluted with ethyl acetate and washed with water. The organic phase was dried over MgSO4, filtered and the solvent is evaporated under vacuum. The final compound is purified by preparative HPLC to give the expected compound.
Method L
Figure imgf000068_0002
L. I Nucleophilic aromatic substitution (general method)
Figure imgf000068_0003
[00297] Cyclopropanecarboxylic acid {5-[4-(6-chloro-pyridin-3-ylmethoxy)-phenyl]-
[l,2,4]triazolo[l,5-a]pyridin-2-yl} -amide prepared by method C (1 eq), an appropriate amine, (1.5 eq.) are mixed in DMSO in a sealed tube. The reaction is heated at 100 0C for 24 hours. Once all the SM disappeared by LCMS, water is added to the reaction mixture and the organics is extracted with ethyl acetate. The organic layer is dried over MgSθ4 and evaporater under vacuum. The final compound is isolated by preparative HPLC. Analytical: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350)
[00298] Preparative HPLC: Waters XBridge Prep Cl 8 5μm ODB 19mm ID x 100mm L (Part
No.186002978). All the methods are using MeCN/H2O gradients. H2O contains either 0.1% TFA or 0.1% NH3.
Method M
Figure imgf000069_0001
M.I Suzuki reaction (general meth nocd d))
Figure imgf000069_0002
[00299] The boronic acid (2eq.) is added to a solution of cyclopropanecarboxylic acid {5-[4-(6- chloro-pyridin-3-ylmethoxy)-phenyl]-[l,2,4]triazolo[l,5-a]pyridin-2-yl} -amide (prepared by method B) in 1.4-dioxan/water (5:1). K2CO3 (2 eq.) and PdCl2dppf (5%) are added to the solution. The resulting mixture is then heated in a microwave at 140 0C for 30 min (This reaction can also be carried out by traditional heating in an oil bath at 900C for 16h under N2). Water is added and the solution is extracted with ethyl acetate. The organic layers are dried over MgSθ4 and evaporated in vacuo. The final compound is obtained after purification by preparative HPLC
Method O
General procedure for the preparation ofsulfones
Figure imgf000070_0001
wherein R4b, R4c and R3b are as described herein.
[00300] A solution of 4-bromobenzenesulfonyl chloride 1 (1.0 g, 3.96 mmol, 1.0 equiv.), sodium sulfite (0.6 g, 4.35 mmol, 1.1 equiv.) and sodium hydrogen carbonate (1.7 g, 79.8 mmol, 5.0 equiv.) in water (10 mL) is heated to 1000C for 4 hours. The reaction mixture becomes clear and the appropriate halide (4.76 mmol, 1.2 equiv.) is added at 1000C. The mixture is stirred at this temperature for 16 hours. The reaction mixture is cooled to room temperature. Then, additional water is added (50 mL) and the resulting aqueous layer is extracted with dichloromethane (3 x 5OmL). The combined organic layers are dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting crude product is purified by chromatography over silica gel to afford the expected sulfone.
Figure imgf000070_0002
wherein R4b, R4c and R3b and nl are as described herein.
[00301] In a microwave vessel, a solution of the sulfone obtained according to the procedure described above (0.84 mmol, 1.5 equiv.), di(pinacolato)diborane (283.0 mg, 1.11 mmol, 2.0 equiv.) potassium acetate (109.0 mg; 1.11 mmol, 2.0 equiv.) in dioxane (2.0 mL) is flushed with argon (3 times). [l,r-Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (20.0 mg, 0.03 mmol, 0.05 equiv.) is then added and the reaction mixture is flushed again with argon (3 times) and heated up to 900C for 20 hours until the reaction is complete on TLC.
[00302] Then, cyclopropanecarboxylic acid (5-bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide
(200.0 mg, 0.56 mmol, 1.0 equiv.), sodium hydrogen carbonate (233.0 mg, 2.78 mmol, 5.0 equiv.), [1,1'- Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (20.0 mg, 0.03 mmol, 0.05 equiv.) and dioxane/water 2: 1 (1.5 mL) are added to the mixture. The reaction mixture is then submitted several times (1-5 times) to microwave irradiations (P: 150 W, T=120°C, t=15 min.) until complete consumption of the starting material 133. Sodium sulfate is added to the reaction mixture (2.0 g) before diluting the latter with dichloromethane (3.0 mL). Purification by chromatography on silica gel (dichloromethane/methanol, 99:1 → 90:10) followed by trituration of the collected compound in methanol affords the expected product with a satisfactory HPLC purity.
Method P
Reaction of alkylation
Figure imgf000071_0001
[00303] The appropriate alkylating agent (1.5eq.) is added to a solution of the acetamide derivative (leq.) obtained by method A and NaH (2eq.) in DMF at 00C. The mixture is stirred for 16hrs at room temperature. The solution is then diluted in water at 00C and the solution is extracted with EtOAc. The organic phases are dried over MgSθ4, filtered and the solvent is removed under vacuum. The final compound is isolated by preparative HPLC.
Method Q
Figure imgf000071_0002
[00304] 2-(4-Bromomethyl-phenyl)-4,4,5,5-tetramethyl-[l,3,2]dioxaborolane (leq.) and K2CO3
(2 eq) (or AgCO3) are dissolved in DMF under N2 and the appropriate phenol (2 eq) is added dropwise. The resulting suspension is heated at 500C for 16h. After this time, the reaction is complete. The compound is extracted with EtOAc and water, washed with brine and dried over MgSO^ Organic layers are filtered and evaporated to afford the desire compound without further purification.
Figure imgf000071_0003
The title compound is optained by method A using the intermediate boronate ester described above.
Method U
Figure imgf000072_0001
[00305] EDCI (1.5 eq.), HOBt (1.5 eq.) and Et3N (2eq.) are added to a solution of {4-[2-
(cyclopropanecarbonyl-amino)-[l,2,4]triazolo[l,5-a]pyridin-5-yl]-phenyl}-acetic acid in DCM at room temperature. The resulting mixture is stirred for 2h at room temperature. An appropriate amine is added to the solution and the reaction is stirred for 16 hrs. Water is added to the reaction mixture, and the later is extracted with EtOAc. The organic phases are dried over MgSθ4, filtered and evaporated under vacuum. Purification by flash chromatography affords the expected product.
Synthesis of representative compounds of the invention Compound 1
[00306] This compound was prepared via Method A using l-methyl-4-[4-(4,4,5,5-tetramethyl-
[l,3,2]dioxaborolan-2-yl)-phenyl]-piperazine.
Compound 2
[00307] This compound was prepared via Method A using 3-(4-morpholinomethyl)- phenylboronic acid pinacol ester hydrochloride.
Compound 3
[00308] This compound was prepared via Method A using 2-(piperidin-l-yl)pyridine-5-boronic acid pinacol ester.
Compound 4
[00309] This compound was prepared via Method A using 2-(pyrrolidin-l-yl)pyrimidine-5- boronic acid pinacol ester.
Compound 5
[00310] This compound was prepared via Method A using 2-(4-methylpiperazin-l-yl)pyridine-
4-boronic acid pinacol ester. Compound 6
[00311] This compound was prepared via Method A using 2-(4-morpholino)pyridine-5-boronic acid pinacol ester.
Compound 7
[00312] This compound was prepared via Method A using biphenyl-4-boronic acid.
Compound 8
[00313] This compound was prepared via Method A using 2-(4-morpholino)pyrimidine-5- boronic acid pinacol ester.
Compound 9
[00314] This compound was prepared via Method A using 2-(piperidin-l-yl)pyrimidme-5- boronic acid pinacol ester.
Compound 10
[00315] This compound was prepared via Method A using 4-benzoylphenylboronic acid.
Compound 11
[00316] This compound was prepared via Method A using [4-
(cyclopropylaminocarbonyl)phenyl]boronic acid.
Compound 12
[00317] This compound was prepared via Method A using 4-benzyloxyphenylboronic acid.
Compound 13
[00318] This compound was prepared via Method A using 4-(N- cyclopropylsulfonamide)phenylboronic acid pinacol ester.
Compound 14
[00319] This compound was prepared via Method A using 3-benzyloxyphenylboronic acid.
Compound 15
[00320] This compound was prepared via Method A using 4-benzyloxy-3-fluorophenylboronic acid. Compound 16
[00321] This compound was prepared via Method A using 2-benzyloxyphenylboronic acid.
Compound 17
[00322] This compound was prepared via Method A using piperidin-l-yl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-methanone.
Compound 18
[00323] This compound was prepared via Method A using 4-[4-(4,4,5,5-tetramethyl-
[l,3,2]dioxaborolan-2-yl)-benzyl]-morpholine.
Compound 19
[00324] This compound was prepared via Method A using pyrrolidin-l-yl-[4-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenyl]-methanone.
Compound 20
[00325] This compound was prepared via Method A using 4-(2-thienyl)phenylboronic acid.
Compound 21
[00326] This compound was prepared via Method D using benzoyl chloride.
Compound 22
[00327] This compound was prepared via Method D using 4-trifluoromethyl-benzoyl chloride.
Compound 23
[00328] This compound was prepared via Method D using phenyl-acetyl chloride.
Compound 24
[00329] This compound was prepared via Method B using morpholine.
Compound 25
[00330] This compound was prepared via Method B using 4-amino-pyridine.
Compound 26
[00331] This compound was prepared via Method B using amino-cyclohexane. Compound 27
[00332] This compound was prepared via Method B using 4-tert-butyl-piperidine.
Compound 28
[00333] This compound was prepared via Method B using [l,4]diazepane.
Compound 29
[00334] This compound was prepared via Method B using 3-fluoro-benzylamine.
Compound 30
[00335] This compound was prepared via Method B using N-methylaniline.
Compound 31
[00336] This compound was prepared via Method B using (4-methoxy-benzyl)-methyl-amine.
Compound 32
[00337] This compound was prepared via Method B using l-methyl-piperidin-4-ylamine.
Compound 33
[00338] This compound was prepared via Method D using 4-fluoro-sulfonylchloride.
Compound 34
[00339] This compound was prepared via Method D using 2-fluorobenzoylchloride.
Compound 35
[00340] This compound was prepared via Method D using pyrazine-2-carbonyl chloride.
Compound 36
[00341] This compound was prepared via Method C using 3-bromomethyl-pyridine hydrobromide.
Compound 37
[00342] This compound was prepared via Method C using 2-bromomethyl-pyridine hydrobromide. Compound 38 [00343] This compound was prepared via Method C using 3-(trifluoromethoxy)benzyl bromide.
Compound 39
[00344] This compound was prepared via Method C using (bromomethyl)cyclobutane.
Compound 40
[00345] This compound was prepared via Method C using iodocyclopentane.
Compound 41
[00346] This compound was prepared via Method C using (bromomethyl)cyclohexane.
Compound 42
[00347] This compound was prepared via Method B using C-pyridin-3-yl-methylamine.
Compound 43
[00348] This compound was prepared via Method B using aniline.
Compound 44
[00349] This compound was prepared via Method D using benzoyl chloride.
Compound 45
[00350] This compound was prepared via Method D using cyclohexanecarbonyl chloride.
Compound 46
[00351] This compound was prepared via Method A using 4-phenoxyphenylboronic acid.
Compound 47
[00352] This compound was prepared via Method G using phenyl boronic acid.
Compound 48
[00353] This compound was prepared via Method C using 3-(chloromethyl)-l-methyl-lH- pyrazole.
Compound 49 [00354] This compound was prepared via Method C using 4-(chloromethyl)-3,5- dimethylisoxazole.
Compound 50
[00355] This compound was prepared via Method C using 5-(chloromethyl)-l,3-dimethyl-lH- pyrazole.
Compound 51
[00356] This compound was prepared via Method C using 4-(2-bromoethyl)-3,5-dimethyl-lH- pyrazole.
Compound 52
[00357] This compound was prepared via Method C using 3-(bromomethyl)-5-methylisoxazole.
Compound 53
[00358] This compound was prepared via Method D using 3-methoxy-benzoyl chloride.
Compound 54
[00359] This compound was prepared via Method D using 2-fluoro-phenyl sulfonyl chloride.
Compound 55
[00360] This compound was prepared via Method D using Pyridine-2-carboxylic acid (acid chloride formed by reaction of oxalyl chloride).
Compound 56
[00361] This compound was prepared via Method B using benzylamine.
Compound 57: Cyclopropanecarboxylic acid [5-(6-benzyloxy-pyridin-3-yl)-[ 1 , 2, 4]triazolo[l, 5- a] pyridin-2-yl] -amide
Figure imgf000077_0001
[00362] At 00C and under N2 atmosphere, benzyl alcohol (2 eq) in a solution of THF was treated with NaH 60 % in mineral oil (4 eq) for 30 min. Then cyclopropanecarboxylic acid [5-(6-chloro-pyridin- 3-yl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide prepared by methaod A was added to the solution and the mixture was stirred at 700C for 3 hours. The reaction was completed. The reaction mixture was quenched with water and the compound was extracted with EtOAc. The compound was washed with brine, dried on MgSO/i, filtrated and concentrated. Compound was purified on Prep HPLC.
Compound 58
[00363] This compound was prepared via Method B using 1,2,3,4-tetrahydro-isoquinoline.
Compound 59
[00364] This compound was prepared via Method D using cyclopropanesulfonyl chloride.
Compound 60
[00365] This compound was prepared via Method B using 2-phenoxy-ethylamine.
Compound 61
[00366] This compound was prepared via Method H using pyrazine.
Compound 62
[00367] This compound was prepared via Method C using 3-(chloromethyl)-l,5-dimethyl-lH- pyrazole.
Compound 63
[00368] This compound was prepared via Method C using 4-(chloromethyl)-2,5-dimethyl-l,3- oxazole.
Compound 64
[00369] This compound was prepared via Method D using pyridine-3-sulfonyl chloride.
Compound 65
[00370] This compound was prepared via Method D using l,3-dimethyl-lH-pyrazole-4-sulfonyl chloride.
Compound 66 [00371] 3-Pyridineboronic acid (1.1 eq.) was added to a solution of cyclopropanecarboxylic acid
[5-(4-bromo-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide, prepared by Method A in 1,4- dioxane/water (5 : 1 ) . K2CO3 (2 eq.) and Pd(dppf)Cl2 (0.03 eq.) (dppf = 1,1 '- Bis(diphenylphosphino)ferrocene) were added to the solution. The resulting mixture was then heated in a sealed tube at 900C for 16hrs. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The final compound was obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350).
Compound 67
[00372] lH-Pyrazole-4-boronic acid (1.1 eq.) was added to a solution of cyclopropanecarboxylic acid [5-(4-bromo-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide, prepared by method A in 1,4- dioxane/water (5 : 1 ) . K2CO3 (2 eq.) and Pd(dppf)Cl2 (0.03 eq.) (dppf = 1,1 '- Bis(diphenylphosphino)ferrocene) were added to the solution. The resulting mixture was then heated in a sealed tube at 900C for 16hrs. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The final compound was obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350).
Compound 68
[00373] 3-Pyridineboronic acid (1.1 eq.) was added to a solution of cyclopropanecarboxylic acid
[5-(6-chloro-pyridin-3-yl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide, prepared by method A in 1,4- dioxane/water (5 : 1 ) . K2CO3 (2 eq.) and Pd(dppf)Cl2 (0.03 eq.) (dppf = 1,1 '- Bis(diphenylphosphino)ferrocene) were added to the solution. The resulting mixture was then heated in a sealed tube at 900C for 16hrs. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The final compound was obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH Cl 8 1.7μm, 2.1mm ID x 50mm L (Part No.186002350).
Compound 69
[00374] This compound was prepared via Method D using 2-methyl-5-propyl-2H-pyrazole-3- carboxylic acid which was reacted first in presence of oxalyl chloride to obtain the acid chloride.
Compound 70
[00375] This compound was prepared via Method D using cyclobutanecarbonyl chloride. Compound 71 [00376] This compound was prepared via Method B using N-methyl pyrazine.
Compound 72
[00377] This compound was prepared via Method E using benzaldehyde.
Compound 73
[00378] This compound was prepared via Method B using 4-ethoxymethyl-piperidine.
Compound 74
[00379] This compound was prepared via Method B using phenyl-piperidin-4-yl-methanone.
Compound 75
[00380] This compound was prepared via Method B using l-benzyl-[l,4]diazepane.
Compound 76
[00381] This compound was prepared via Method D using 2-phenyl-ethanesulfonyl chloride.
Compound 77
[00382] This compound was prepared via Method D using Phenyl-methanesulfonyl chloride.
Compound 78
[00383] This compound was prepared via Method C using 3-(chloromethyl)-6-
(trifluoromethyl)pyridine.
Compound 79
[00384] This compound was prepared via Method D using phenylsulfonyl chloride.
Compound 80
[00385] This compound was prepared via Method B using 2-pyrrolidin-l-yl-ethylamine.
Compound 81
[00386] This compound was prepared via Method B using 3-morpholin-4-yl-propylamine.
Compound 82
[00387] This compound was prepared via Method H using 4-phenethyl-piperidine. Compound 83
[00388] This compound was prepared via Method H using 4-(4-chloro-phenyl)-piperidine.
Compound 84
[00389] This compound was prepared via Method H using 3-phenylpiperidine.
Compound 85
[00390] This compound was prepared via Method D using 4-propyl-benzenesulfonyl chloride.
Compound 86
[00391] This compound was prepared via Method B using 4-(4-chloro-phenyl)-piperidine.
Compound 87
[00392] This compound was prepared via Method B using phenethylamine.
Compound 88
[00393] This compound was prepared via Method D using 2-(3-fluoro-phenyl)-ethanesulfonyl chloride.
Compound 89
[00394] This compound was prepared via Method B using piperidine.
Compound 90
[00395] This compound was prepared via Method B using butyl-(3-morpholin-4-yl-propyl)- amine.
Compound 91
[00396] This compound was prepared via Method C using 4-(chloromethyl)-2-[4-
(trifluoromethyl)phenyl]-thiazole.
Compound 92
[00397] This compound was prepared via Method C using 4-acetamidobenzyl chloride.
Compound 93
[00398] This compound was prepared via Method H using 2-(tetrahydro-pyran-4-yl)-ethylamine. Compound 94
[00399] This compound was prepared via Method B using 4-(2-piperidin-4-yl-ethyl)- morpholine.
Compound 95
[00400] This compound was prepared via Method H using 4-(2-piperidin-4-yl-ethyl)- morpholine.
Compound 96
[00401] This compound was prepared via Method B using methyl-phenethyl-amine.
Compound 97
[00402] This compound was prepared via Method B using 2-(4-trifluoromethyl-phenyl)- ethylamine.
Compound 98
[00403] This compound was prepared via Method C using l-(2-bromo-ethyl)-lH-pyrazole.
Compound 99
[00404] This compound was prepared via Method C using 3-(chloromethyl)-l,2,4-oxadiazole.
Compound 100
[00405] This compound was prepared via Method B using 2-phenoxyethylamine.
Compound 101
[00406] This compound was prepared via Method B using 4-piperidin-4-yl-morpholine.
Compound 102
[00407] This compound was prepared via Method B using 1 , 1 -dimethyl^-morpholin^-yl- ethylamine.
Compound 103
[00408] This compound was prepared via Method B using benzyl-methyl-piperidin-4-yl-amine.
Compound 104 [00409] This compound was prepared via Method H using benzylamine.
Compound 105
[00410] This compound was prepared via Method B using [2-(3,4-dimethoxy-phenyl)-ethyl]- methyl-amine.
Compound 106
[00411] This compound was prepared via Method B using 2-(l-phenyl-lH-pyrazol-4-yl)- ethylamine.
Compound 107
[00412] This compound was prepared via Method A using 1 -benzyl- lH-pyrazole-4-boronic acid.
Compound 108
[00413] This compound was prepared via Method D using 3-phenoxy-propionyl chloride.
Compound 109
[00414] This compound was prepared via Method D using 3-phenyl-propionyl chloride.
Compound 110
[00415] This compound was prepared via Method B using 3-(piperidin-4-yloxy)-pyridine.
Compound 111
[00416] This compound was prepared via Method B using phenethylamine.
Compound 112
[00417] This compound was prepared via Method H using 4-phenethyl-piperidine.
Compound 113
[00418] This compound was prepared via Method B using l-(3-chloro-phenyl)-piperazine.
Compound 114
[00419] This compound was prepared via Method B using 3 -phenyl-propylamine.
Compound 115 [00420] This compound was prepared via Method C using l-(2-bromoethoxy)-4-fluorobenzene.
Compound 116
[00421] This compound was prepared via Method C using N-(chloroacetyl)-3-fluoroaniline.
Compound 117
[00422] This compound was prepared via Method C using l-(4-benzyl-piperidin-l-yl)-2-chloro- ethanone.
Compound 118
[00423] This compound was prepared via Method C using 2-chloro-N-methyl-N-phenyl- acetamide.
Compound 119
[00424] This compound was prepared via Method B using (S)-I -benzyl-pyrrolidin-3-ylamine.
Compound 120
[00425] This compound was prepared via Method B using (R)-I -benzyl-pyrrolidin-3-ylamine.
Compound 121: Cyclopropanecarboxylic acid [5-(l -benzyl- lH-indol- 5 -yl)-[ '1,2,4) 'triazolo[ 1,5- a] pyridin-2-yl] -amide
Figure imgf000084_0001
121.1 l-Benzyl-5-(4, 4, 5, 5-tetramethyl-fl, 3, 2]dioxaborolan-2-yl)-lH-indole
Figure imgf000084_0002
[00426] To a solution of 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-indole (2.27 mmol; 1.0 equiv.) in acetone (10.0 mL) at room temperature were added under argon benzyl bromide (3.18 mmol; 1.4 aquiv.) and cesium carbonate (3.18 mmol; 1.4 equiv.). The reaction mixture was heated for 4 hours at reflux. The mixture was then cooled to room temperature, quenched by addition of a saturated aqueous solution of sodium hydrogen carbonate (100 mL) and extracted with dichloromethane (2 x 100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash chromatography over silica gel (dichloromethane/ethyl acetate) to afford the expected boronate as a white solid used in the next step without further purification.
121.2 Cyclopropa neca rboxylic acid [5-(l-benzyl-lH-indol-5-yl)-[l,2,4]triazolo[l,5- a] pyridin-2-yl] -amide
Figure imgf000085_0001
[00427] The title compound was then synthesised by Method A using l-benzyl-5-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-indol.
Compound 122
[00428] This compound was prepared via Method D using 2-phenoxy-ethanesulfonyl chloride.
Compound 123
[00429] This compound was prepared via Method H using phenethylamine.
Compound 124
[00430] This compound was prepared via Method B using 2-pyridin-3-yl-ethylamine.
Compound 125
[00431] This compound was prepared via Method C using the mesylate derivative of (4- pyrazol- 1 -yl-phenyl)-methanol.
Compound 126
[00432] This compound was prepared via Method C using the mesylate derivative of [4-(4- methyl-piperazin- 1 -ylmethyl)-phenyl] -methanol. Compound 127
[00433] This compound was prepared via Method C using 3-(2-chloro-ethyl)-pyridine.
Compound 128
[00434] This compound was prepared via Method B using 2-piperidin-4-yl-lH-benzoimidazole.
Compound 129
[00435] This compound was prepared via Method A using 4-[4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl]morpholine.
Compound 130
[00436] This compound was prepared via Method I using N-methyl-pyrazine.
Compound 131
[00437] This compound was prepared via Method B using 4-(2-methoxy-phenyl)-piperidine.
Compound 132
[00438] This compound was prepared via Method B using 4-(4-chloro-phenyl)-piperidin-4-ol.
Compound 133
[00439] This compound was prepared via Method B using 4-o-tolyloxymethyl-piperidine.
Compound 134
[00440] This compound was prepared via Method B using 3, 5-dimethyl piperidine.
Compound 135
[00441] This compound was prepared via Method B using N-(4-piperidin-l-ylbenzyl)propan-2- amine.
Compound 136
[00442] This compound was prepared via Method B using propyl-(tetrahydro-furan-2-ylmethyl)- amine.
Compound 137
[00443] This compound was prepared via Method B using 4-fluoropiperidine. Compound 138
[00444] This compound was prepared via Method B using 2-piperidin-4-yl-lH-indole.
Compound 139
[00445] This compound was prepared via Method I using piperidine.
Compound 140
[00446] This compound was prepared via Method I using piperidine-4-carboxylic acid amide.
Compound 141
[00447] This compound was prepared via Method I using 1-piperazin-l-yl-ethanone.
Compound 142
[00448] This compound was prepared via Method I using l-pyridin-2-yl-piperazine.
Compound 143
[00449] This compound was prepared via Method I using 2-piperazin-l-yl-pyrimidine.
Compound 144
[00450] This compound was prepared via Method B using 2-methylpiperidine.
Compound 145
[00451] This compound was prepared via Method B using 3-methylpiperidine.
Compound 146
[00452] This compound was prepared via Method B using 4-methyl piperidine.
Compound 147
[00453] This compound was prepared via Method B using 4-phenethyl-piperidine.
Compound 148
[00454] This compound was prepared via Method B using 4-trifloromethylpiperidine.
Compound 149 [00455] This compound was prepared via Method B using 6-fluoro-3-piperidin-4-yl- benzo[d]isoxazole.
Compound 150
[00456] This compound was prepared via Method B using N'-benzyl-N,N- dimethylethylenediamine.
Compound 151
[00457] This compound was prepared via Method B using (4-fluoro-benzyl)-(2-methoxy-l- methyl-ethyl)-amine hydrochloride.
Compound 152
[00458] This compound was prepared via Method B using l-piperidin-4-yl-lH-benzotriazole.
Compound 153
[00459] This compound was prepared via Method B using (4-fluoro-benzyl)-(tetrahydro-furan-
2-ylmethyl)-amine.
Compound 154
[00460] This compound was prepared via Method B using 4-[2-(2-methyl-imidazol-l-yl)-ethyl]- piperidine.
Compound 155
[00461] This compound was prepared via Method I using methyl-phenethyl-amine.
Compound 156
[00462] This compound was prepared via Method B using 4-benzyl-piperidin-4-ol.
Compound 157
[00463] This compound was prepared via Method E using benzyl bromide.
Compound 158: N-(5-(4-((lH-tetrazol-5-yl)methyllH-tetrazol-5-yl)methoxy)phenyl)-
[1, 2, 4]triazolo[l, 5-a]pyridin-2-yl)cyclopropanecarboxamide
Figure imgf000089_0001
[00464] A solution of cyclopropanecarboxylic acid [5-(4-cyanomethoxy-phenyl)-
[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide (1 equiv), sodium azide (2 equiv), and ammonium chloride (2 equiv) was stirred at 00C under nitrogen for 30 minutes. Afterwards the mixture was subjected to 1000C for 16 hours. The crude of the reaction was diluted with ethyl acetate and washed with water. The organic phase was dried over MgS O4, filtered, and dried under vacuum. The crude was diluted with DMSO and submitted for preparative HPLC purification: UPLC system (XBridge™ Prep Cl 8, 5 μm, 19x100 mm column); 8 min LC; flow: 20 mL/min; gradient: from 30% to 70% acetonitrile in water 0.1 % TFA; to afford the final pure product (10% yield).
Compound 159
[00465] This compound was prepared via Method I using methyl-(4-pyridin-2-yl-benzyl)-amine.
Compound 160
[00466] This compound was prepared via Method I using (l,5-dimethyl-lH-pyrazol-3- ylmethyl)-methyl-amine.
Compound 161
[00467] This compound was prepared via Method I using methyl-(4-pyrimidin-5-yl-benzyl)- amine.
Compound 162
[00468] This compound was prepared via Method I using methyl-pyridin-3-ylmethyl-amine.
Compound 163
[00469] This compound was prepared via Method F using 2-(4-benzyloxy-phenyl)-4,4,5,5- tetramethyl- [ 1 ,3 ,2] dioxaborolane.
Compound 164
[00470] This compound was prepared via Method B using 3-trifluoromethyl piperidine. Compound 165
[00471] This compound was prepared via Method L using morpholine.
Compound 166
[00472] This compound was prepared via Method M using 4-(4,4,5,5-tetramethyl-
[l,3,2]dioxaborolan-2-yl)-lH-pyrazole.
Compound 167
[00473] This compound was prepared via Method L using N-methyl morpholine.
Compound 168
[00474] This compound was prepared via Method B using 4,4-difluoro-piperidine.
Compound 169
[00475] This compound was prepared via Method B using 3-phenyl-piperidine.
Compound 170
[00476] This compound was prepared via Method E using C-pyridin-3-yl-methylamine.
Compound 171
[00477] This compound was prepared via Method E using C-pyridin-2-yl-methylamine.
Compound 172
[00478] This compound was prepared via Method E using 2-pyridin-3-yl-ethylamine.
Compound 173
[00479] This compound was prepared via Method E using C-(l,5-dimethyl-lH-pyrazol-3-yl)- methylamine.
Compound 174
[00480] This compound was prepared via Method L using pirrolidine.
Compound 175
[00481] This compound was prepared via Method B using 3,3-dimethyl-piperidine. Compound 176: N-(5-(4-((6-cyanopyridin-3-yl)methoxy)phenyl)-[ 1 ,2,4]triazolo[l ,5-a] pyridin-2- yl)cyclopropanecarboxamide
176.1: Sy n th es is of 5-[4-(4,4,5,5-Tetramethyl-[l,3,2Jdioxaborolan-2-yl)-phenoxymethylJ- pyridine-2-carbonitrile
Figure imgf000091_0001
[00482] To 4-hydroxyphenylboronic acid pinacol ester (25 g; O.l lmol; 1.0 equiv.) in acetone
(250 mL) at room temperature were added under argon 5-chlomomethyl-pyridine-2-carbonitrile (19 g; 0.12 mol; 1.1 equiv.) and cesium carbonate (73.9 g, 0.22 mol; 2 equiv.). The reaction mixture was heated for 4 hours at reflux. The mixture was then cooled to room temperature, the acetone was evaporated. Water (200 mL) was added and the product was extracted with EtOAc (3 x 200 mL). The organic layer was dried over magnesium sulfate, filtered and concentrated to dryness. The resulting residue was purified by chromatography over silica gel (petrol: EtOAc 10:1) to afford the expected boronate as a white solid.
176.2: Synthesis of Cyclopropanecarboxylic acid (5-[4-(6-cyano-pyridin-3-ylmethoxy)- phenyl]-[l,2, 4]triazolo[l, 5-aj 'pyridin-2-yl} -amide
Figure imgf000091_0002
[00483] 5-[4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-phenoxymethyl]-pyridine-2- carbonitrile (10 g, 0.03 mol, 1.1 equiv.) was added to a solution of cyclopropanecarboxylic acid (5- bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide (7.6 g, 0.027 mol) in 1,4-dioxane/water (4:1; 70 mL). K2CO3 (7.45, 0.054 mol, 2 eq.) and PdCl2dppf (5%) were added to the solution. The resulting mixture was then heated in an oil bath at 900C for 4 to 16h under N2 until completion (monitored by LCMS). 1,4-Dioxane was removed under vacuum, and water/EtOAc were added and the solid was filtered. The obtained solid was dissolved in methanol/DCM, dried over MgSθ4 and the final compound was obtained after purification by flash chromatography, eluted with neat EtOAc Compound 177
[00484] This compound was prepared via Method D using 1 -cyano-cyclopropanecarbonyl chloride.
Compound 178
[00485] This compound was prepared via Method M using l-methyl-4-(4,4,5,5-tetramethyl-
[l,3,2]dioxaborolan-2-yl)-lH-pyrazole.
Compound 179: Cyclopropanecarboxylic acid {5-[l-(3-phenyl-propionyl)-lH-indol-5-yl]-
[1, 2, 4]triazolo[l, 5-aj ' pyridin-2-yl} -amide
179.1 3-Phenyl-l-[5-(4, 4, 5, 5-tetramethyl-[l, 3, 2] dioxaborolan-2-yl)-indol-l-yl] -propan-l-one
Figure imgf000092_0001
[00486] To a solution of 5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-indole (2.27 mmol; 1.0 equiv.) in DMF (10.0 mL) at room temperature were added under argon 3-phenyl-propionyl chloride (3.18 mmol; 1.4 aquiv.) and sodium hydride (3.18 mmol; 1.4 equiv.). The reaction mixture was heated at 600C for 16 hrs. The mixture was allowed to cool to room tempretaure and was quenched by addition of water (100 mL) and extracted with dichloromethane (2 x 100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash chromatography over silica gel (dichloromethane/ethyl acetate) to afford the expected boronate as a white solid used in the next step without further purification.
179.2 Cy c lop rop a n e c a rb oxy l i c a c i d { 5-[l-(3-phenyl-propionyl)-lH-indol-5-yl]-
[1, 2, 4]triazolo[l, 5 -a] pyridin-2-yl} -amide
Figure imgf000092_0002
[00487] The title compound was prepared by Method A using 3-phenyl-l-[5-(4,4,5,5- tetramethyl-[l,3,2]dioxaborolan-2-yl)-indol-l-yl]-propan-l-one.
Compound 180
[00488] This compound was prepared via Method D using Benzenesulfonyl chloride.
Compound 181
181.1 Preparation of phenethylamino-acetonitrile
[00489] Chloro-acetonitrile (1.5) eq was added to a solution of phenethylamine (leq) and K2CO3
(2 eq) in CH3CN. The mixture was stirred at 6O0C for 4hrs. After completion of the reaction, the mixture was filtered and the filtrate was concentrated under reduced pressure. Purification by flash chromatography gave the expected compound.
181.2 Compound 181 was prepared using Method B using phenethylamino-acetonitrile.
Compound 182
[00490] This compound was prepared via Method E using 3-bromomethyl-pyridine.
Compound 183: Cyclopropanecarboxylic acid (5-{4-[6-(2H-tetrazol-5-yl)-pyridin-3-ylmethoxy]- phenyl}-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide
Figure imgf000093_0001
[00491] A solution of cyclopropanecarboxylic acid {5-[4-(6-cyano-pyridin-3-ylmethoxy)- phenyl]-[l,2,4]triazolo[l,5-a]pyridin-2-yl}-amide (143 mg, 0.35 mmol, 1 equiv), sodium azide (46 mg, 0.7 mmol, 2 equiv), ammonium chloride (38 mg, 0.7 mmol, 2 equiv) was stirred at 00C under nitrogen for 30 minutes. Afterwards the mixture was subjected for 1000C for 16 hours. The crude of the reaction was diluted with ethyl acetate and washed with water. The organic phase was dried over MgSO/i, filtered, and dried under vacuum. The crude was diluted with DMSO and submitted for preparative HPLC purification: UPLC system (XBridge™ Prep C 18, 5 μm, 19x100 mm column); 8 min LC; flow: 20 mL/min; gradient: from 30% to 70% acetonitrile in water 0.1 % TFA; to afford the final pure product (9 mg, 6% yield).
Compound 184
[00492] This compound was prepared via Method D using phenyl-methanesulfonyl chloride.
Compound 185: Cyclopropanecarboxylic acid [5-(l-pyridin-3-ylmethyl-lH-pyrazol-4-yl)-
[1, 2, 4]triazolo[l, 5-a]pyridin-2-yl] -amide
185.1 3-[4-(4, 4, 5, 5-Tetramethyl-[ 1 , 3, 2] dioxaborolan-2-yl)-pyrazol-l-ylmethyl] -pyridine
Figure imgf000094_0001
[00493] To a solution of 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (2.27 mmol; 1.0 equiv.) in acetone (10.0 mL) at room temperature were added under argon 3-Chloromethyl- pyridine hydrochloride (3.18 mmol; 1.4 aquiv.) and cesium carbonate (2.8 equiv.). The reaction mixture was heated for 4 hours at reflux. The mixture was then cooled to room temperature, quenched by addition of a saturated aqueous solution of sodium hydrogen carbonate (100 mL) and extracted with dichloromethane (2 x 100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash chromatography over silica gel (dichloromethane/ethyl acetate) to afford the expected boronate as a white solid used in the next step without further purification.
185.2 Cy c l op rop a n e c a r b o xy l i c a c i d [ 5-(l-pyridin-3-ylmethyl-lH-pyrazol-4-yl)-
[1, 2, 4]triazolo[l, 5-a] pyridin-2-yl] -amide
Figure imgf000094_0002
[00494] The title compound was prepared by Method A using 3-[4-(4,4,5,5-tetramethyl-
[l,3,2]dioxaborolan-2-yl)-pyrazol-l-ylmethyl]-pyridine. Compound 186: Cyclopropanecarboxylic acid {5-[l-(3-phenyl-propionyl)-lH-pyrazol-4-ylJ-
[1, 2, 4]triazolo[l, 5-a]pyridin-2-yl}-amide
186.1 3-Phenyl-l-[4-(4, 4, 5, 5-tetramethyl-fl, 3, 2] dioxaborolan-2-yl)-pyrazol-l-yl] -propan-1- one
Figure imgf000095_0001
[00495] To a solution of 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH-pyrazole (2.27 mmol; 1.0 equiv.) in DMF (10.0 mL) at room temperature were added under argon 3-phenyl-propionyl chloride (3.18 mmol; 1.4 aquiv.) and sodium hydride (3.18 mmol; 1.4 equiv.). The reaction mixture was heated at 600C. The mixture was allowed to cool to room temperature and quenched by addition of a saturated aqueous solution of water (100 mL) and extracted with dichloromethane (2 x 100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to dryness. The resulting residue was purified by flash chromatography over silica gel (dichloromethane/ethyl acetate) to afford the expected boronate as a white solid used in the next step without further purification.
186.2 Cyc lop rop a n eca rb oxy li c a c id { 5-[l-(3-phenyl-propionyl)-lH-pyrazol-4-yl]-
[1, 2, 4]triazolo[l, 5-aj ' pyridin-2-yl} -amide
Figure imgf000095_0002
[00496] The title compound was prepared by Method A uising 3-phenyl-l-[4-(4,4,5,5- tetramethyl- [ 1 ,3 ,2] dioxaborolan-2-yl)-pyrazol- 1 -yl] -propan- 1 -one.
Compound 187
[00497] This compound was prepared via Method K using azidomethyl-benzene.
Compound 188
[00498] This compound was prepared via Method K using 5-azidomethyl-2-trifluoromethyl- pyridine. Compound 189
[00499] This compound was prepared via Method C using the metsylate derivative of 1-pyridin-
2-yl-ethanol.
Compound 190
[00500] This compound was prepared via Method C using the mesylate derivative of 6- hydroxymethyl-nicotinic acid methyl ester.
Compound 191
[00501] This compound was prepared via Method D using cyclopropanesulfonyl chloride.
Compound 192: 5-{4-[2-(Cyclopropanecarbonyl-amino)-[l,2,4]triazolo[l,5-a]pyridin-5-yl]- phenoxymethyl}-pyridine-2-carboxylic acid amide
Figure imgf000096_0001
[00502] To a solution of cyclopropanecarboxylic acid {5-[4-(6-cyano-pyridin-3-ylmethoxy)- phenyl]-[l,2,4]triazolo[l,5-a]pyridin-2-yl}-amide (30 mg, 0.073 mmol, 1 equiv) and K2CO3 (10 mg, 0.073 mmol, 1 equiv) in DMSO (0.2 mL) at 100C, 30% H2O2 (17 μL, 0.146 mmol, 2 equiv) was added dropwise. After stirring at room temperature the mixture for 4 h, the mixture was diluted with DMSO and filtered. The filtrate was submitted for preparative HPLC purification: UPLC system (XBridge™ Prep C18, 5 μm, 19x100 mm column); 8 min LC; flow: 20 mL/min; gradient: from 30% to 70% acetonitrile in water 0.1 % TFA; isolating the final product (25 mg, 81% yield).
Compound 193
[00503] This compound was prepared via Method K using 2-azidomethyl-pyridine.
Compound 194
[00504] This compound was prepared via Method O using 2-bromomethyl-pyridine.
Compound 195 [00505] This compound was prepared via Method O using 3-bromomethyl-pyridine.
Compound 196
[00506] This compound was prepared via Method C using 3-chloromethyl-pyridine 1 -oxide.
Compound 197
[00507] This compound was prepared via Method C using 5-chloromethyl-2-methyl-pyridine.
Compound 198
[00508] This compound was prepared via Method C using 2-chloro-5-chloromethyl-pyridine.
Compound 199
[00509] This compound was prepared via Method C using 3 -chloromethyl-1 -methyl- IH-
[l,2,4]triazole.
Compound 200
Figure imgf000097_0001
[00510] 2-(4-Bromomethyl-phenyl)-4,4,5,5-tetramethyl-[l,3,2]dioxaborolane (leq) and DIPEA
(2 eq) are dissolved in DCM/MeOH (5:1 v: v) under N2 and thiomorpholine 1 , 1 -dioxide (2 eq) was added portionwise. The resulting solution was stirred at room temperature for 16h. After this time, the reaction was complete. The solvent was evaporated. The compound was extracted with EtOAc and water, washed with brine and dried over MgS O4. Organic layers were filtered and evaporated. The final compound was isolated without further purification.
Suzuki coupling
Figure imgf000097_0002
[00511] 4-[4-(4,4,5,5-Tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzyl]-thiomorpholine-l,l- dioxide (l.leq.) was added to a solution of cyclopropanecarboxylic acid (5-bromo-[l,2,4]triazolo[l,5- a]pyridin-2-yl)-amidein 1,4-dioxane/water (4:1). K2CO3 (2 eq.) and PdCl2dppf (0.03 eq.) were added to the solution. The resulting mixture was then heated in an oil bath at 900C for 16h under N2. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The final compound was obtained after purification by flash chromatography.
Alternative route to compound 200:
Figure imgf000098_0001
[00512] 4-(Hydroxymethyl)phenylboronic acid (l.leq.) was added to a solution of cyclopropanecarboxylic acid (5-bromo-[l,2,4]triazolo[l,5-a]pyridin-2-yl)-amide in 1,4-dioxane/water (4:1). K2CO3 (2 eq.) and PdCl2dppf (0.03 eq.) were added to the solution. The resulting mixture was then heated in an oil bath at 900C for 16h under N2. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The resulting mixture was used without further purification.
Figure imgf000098_0002
[00513] To a solution of cyclopropanecarboxylic acid [5-(4-hydroxymethyl-phenyl)-
[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide (1.0 eq) in chloroform was slowly added the phosphorus tribromide (1.0 equiv.). The reaction mixture was stirred at room temperature for 20 hours, quenched with ice and water (20 mL) and extracted with dichloromethane. The organic layer was dried over MgSO/i, filtered and concentrated to dryness. The resulting white residue was triturated in dichloromethane/diethyl ether 2:1 (20 mL) to afford the expected product as a white solid.
[00514] Cyclopropanecarboxylic acid [5-(4-bromomethyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-
2-yl]-amide ( l eq) and DIPEA (2 eq) were dissolved in DCM/MeOH (5:1 v:v) under N2 and thiomorpholine 1,1 -dioxide (1.1 eq) was added dropwise. The resulting solution was stirred at room temperature for 16h. After this time, the reaction was complete. The solvent was evaporated. The compound was dissolved in DCM, washed with water and dried over MgSO^ Organic layers were filtered and evaporated. The final compound was isolated by column chromatography using EtOAc to afford the desired product.
Compound 201
[00515] This compound was prepared via Method C using 4-(2-chloro-ethyl)-3,5-dimethyl- isoxazole.
Compound 202:
[00516] This compound was prepared via Method P using 3-bromomethyl-pyridine.
Compound 203
[00517] This compound was prepared via Method B using 6-methoxy-pyridin-3-ylamine.
Compound 204
[00518] This compound was prepared via Method B using 6-morpholin-4-yl-pyridin-3-ylamine.
Compound 205
[00519] This compound was prepared via Method B using 6-(4-methyl-piperazin-l-yl)-pyridin-
3-ylamine.
Compound 206
[00520] This compound was prepared via Method B using pyridin-3-ylamine. Compound 207 [00521] This compound was prepared via Method B using thiomorpholine 1,1 -dioxide.
Compound 208
[00522] This compound was prepared via Method P using 3-bromomethyl-pyridine.
Compound 209
[00523] This compound was prepared via Method B using 4-hydroxy piperidine.
Compound 210
[00524] This compound was prepared via Method B using piperidine-4-carbonitrile.
Compound 211: Cyclopropanecarboxylic acid {5-[4-(2-pyridin-2-yl-ethyl)-phenyl]-[ 1,2,4] triazolo[ 1,5- a]pyridin-2-yl}-amide
Figure imgf000100_0001
[00525] Potassium tert-butoxide (1.3 eq.) was added to an ice-cooled solution of 2-[(triphenyl- λ5-phosphanyl) -methyl] -pyridine (1.1 eq.) in THF (10 mL/mmol). The resulting mixture was stirred for 30 min at 00C, then at room temperature for another 30 min. A solution of cyclopropanecarboxylic acid [8-(4-formyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide (1 eq.) in THF (10 mL/mmol) was added dropwise to the reaction mixture. The stirring was maintained for 8hrs. The reaction was then quenched with water and extracted with ethyl actate. The organic phase was dried aver MgSO/i, filtered and removed under vacuum. The solid was washed with methanol to afford the title compound in 69% yield. [00526] Cyclopropanecarboxylic acid {5-[4-((E)-2-pyridm-3-yl-vinyi)-phenyi]-
[l,2,4]triazolo[l,5-a]pyridin-2-yl} -amide (320 mg, 0.84 mol) was dissolved in 10 mL MeOH. Pd/C (50 mg) was added and the reaction is put under H2 at normal pressure. The reaction was stirred for 2 hrs. The reaction mixture was filtered through Celite®. The organic solvent was removed under pressure. NaHCθ3 saturated solution was added to resulting mixture. The compound was extracted with EtOAc. The organic phase was dried over MgSθ4, filtered and removed under vacuum to afford the title compound in 100% yield. Compound 212
[00527] This compound was prepared via Method J using 4-chloro-2-fluoro-phenylamine.
Compound 213
[00528] This compound was prepared via Method J using 3,3-dimethyl-morpholine.
Compound 214
[00529] This compound was prepared via Method J using cis-2,6-dimethyl-morpholine.
Compound 215
[00530] This compound was prepared via Method B using cis-2,6-dimethyl-morpholine.
Compound 216
[00531] This compound was prepared via Method B using 3,3-dimethyl-morpholine.
Compound 217
[00532] This compound was prepared via Method B using (lS,4S)-2-oxa-5-aza- bicyclo[2.2.1 ]heptanes.
Compound 218
[00533] This compound was prepared via Method B using 5-cyclopropyl-2-methyl-2H-pyrazol-
3-ylamine.
Compound 219
[00534] This compound was prepared via Method B using morpholin-4-yl-piperidin-4-yl- methanone.
Compound 220
[00535] This compound was prepared via Method B using 1-piperazin-l-yl-ethanone.
Compound 221
[00536] This compound was prepared via Method B using pyridazin-3-ylamine.
Compound 222
[00537] This compound was prepared via Method J using pyridazin-3-ylamine. Compound 223
[00538] This compound was prepared via Method J using pyridin-3-ylamine.
Compound 224
[00539] This compound was prepared via Method J using (4-amino-phenyl)-acetonitrile.
Compound 225
[00540] This compound was prepared via Method J using (2-amino-phenyl)-acetonitrile.
Compound 226
[00541] This compound was prepared via Method J using (4-amino-phenyl)-acetonitrile.
Compound 227
[00542] This compound was prepared via Method J using 4-amino-benzamide.
Compound 228
[00543] This compound was prepared via Method J using 3-amino-benzamide.
Compound 229
[00544] This compound was prepared via Method J using pyrimidin-2-ylamine.
Compound 230
[00545] This compound was prepared via Method J using (lS,4S)-2-oxa-5-aza- bicyclo[2.2.1 Jheptane.
Compound 231
[00546] This compound was prepared via Method J using 2-phenyl-morpholine.
Compound 232
[00547] This compound was prepared via Method J using piperidine-4-carbonitrile.
Compound 233
[00548] This compound was prepared via Method J using 4-fluoropiperidine.
Compound 234
[00549] This compound was prepared via Method J using 4,4-difluoropiperidine. Compound 235
[00550] This compound was prepared via Method J using 6-(4-methyl-piperazm-l-yl)-pyridin-3- ylamine.
Compound 236
[00551] This compound was prepared via Method J using 6-methoxy-pyridin-3-ylamine.
Compound 237
[00552] This compound was prepared via Method J using 6-morpholin-4-yl-pyridin-3-ylamine.
Compound 238
[00553] This compound was prepared via Method Q using phenol.
Compound 239: Cy c lop rop a n e c a r b oxy l i e a c i d { 5-[4-(6-cyano-pyridin-3-yl)-phenyl]-
[1, 2, 4]triazolo[l, 5-a]pyridin-2-yl}-amide
Figure imgf000103_0001
[00554] 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-benzonitrile (1.1 eq.) was added to a solution of cyclopropanecarboxylic acid [5-(4-bromo-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide, prepared by method A in 1,4-Dioxane/water (5:1). K2CO3 (2 eq.) and Pd(dppf)Cl2 (0.03 eq.) (dppf = l,l '-Bis(diphenylphosphino)ferrocene) were added to the solution. The resulting mixture was then heated in a sealed tube at 900C for 16hrs. Water was added and the solution was extracted with ethyl acetate. The organic layers were dried over MgSθ4 and evaporated in vacuo. The final compound was obtained after purification by preparative HPLC. Analytical: Waters Acquity UPLC BEH Cl 8 1.7μm, 2. lmm ID x 50mm L (Part No.186002350).
Compound 240
[00555] This compound was prepared via Method J using 4-trifluoromethyl piperidine. Compound 241
[00556] This compound was prepared via Method J using l-(2,2,2-trifluoro-ethyl)-piperazine.
Compound 242
[00557] This compound was prepared via Method J using 4-hydroxy-piperidine.
Compound 243
[00558] This compound was prepared via Method J using 2-piperidin-4-yl-propan-2-ol.
Compound 244
[00559] This compound was prepared via Method J using pyridin-2-ylamine.
Compound 245
[00560] This compound was prepared via Method J using 2,4-difluoro-3-methoxy-phenylamine.
Compound 246
[00561] This compound was prepared via Method J using 2,6-difluoro-phenylamine
Compound 247
[00562] This compound was prepared via Method J using diethyl-piperidin-4-yl-amine.
Compound 248
[00563] This compound was prepared via Method J using 2-fluoro-5-trifluoromethyl- phenylamine.
Compound 249
[00564] This compound was prepared via Method J using 3-amino-4-methyl-benzamide.
Compound 250
[00565] This compound was prepared via Method J using piperidin-4-yl-methanol.
Compound 251
[00566] This compound was prepared via Method Q using 3-hydroxy-benzamide.
Compound 252
[00567] This compound was prepared via Method J using diethyl-pyrrolidin-3-yl-amine. Compound 253
[00568] This compound was prepared via Method J using (lR,4R)-2-ethyl-2,5-diaza- bicyclo[2.2.1 Jheptane.
Compound 254: Cyclopropanecarboxylic acid { 5-[4-(3-oxo-morpholin-4-ylmethyl)-phenyl] -
[1, 2, 4]triazolo[l, 5-aj ' pyridin-2-yl} -amide
Figure imgf000105_0001
254.1 4-[4-(4, 4, 5, 5-Tetramethyl-[l, 3, 2] ' dioxaborolan-2-yl) -benzyl] ' -morpholin-3-one
Figure imgf000105_0002
[00569] NaH (81 mg, 3 eq.) was added to a solution of morpholin-3-one in DCM. 2-(4- bromomethyl-phenyl)-4,4,5,5-tetramethyl-[l,3,2]dioxaborolane was added to the resulting solution and the reaction was stirred at room temperature for 16 hrs. DCM was evaporated, followed by addition of water. The solution was extracted with EtOAc. The organic layers were dried over MgSC^ and evaporated in vacuo to afford the title product used in the next step without further purification.
254.2 Cyc lop rop a n e ca rb oxy li c a c i d { 5-[4-(3-oxo-morpholin-4-ylmethyl)-phenyl]-
[1, 2, 4]triazolo[l, 5-a]pyridin-2-yl}-amide
Figure imgf000106_0001
[00570] The title compound was then synthesized by Method A.
Compound 255
[00571] This compound was prepared via Method J using 3-amino-4-methoxy-benzamide.
Compound 256
[00572] This compound was prepared via Method J using 2-fluoro-6-methyl-pyridin-3-ylamine.
Compound 257
[00573] This compound was prepared via Method J using 3,5-difluoro-pyridin-2-ylamine.
Compound 258
[00574] This compound was prepared via Method J using 4-amino-3-fluoro-benzonitrile.
Compound 259
[00575] This compound was prepared via Method J using 2-fluoro-4-methyl-phenylamine.
Compound 260
[00576] This compound was prepared via Method J using pyrrolidine.
Compound 261
[00577] This compound was prepared via Method J using aniline.
Compound 262
[00578] This compound was prepared via Method J using N-methyl-N-pyrrolidin-3-yl- acetamide.
Compound 263
[00579] This compound was prepared via Method J using dimethyl-pyrrolidin-3-yl-amine. Compound 264
[00580] This compound was prepared via Method J using 3,3-difluoro-pyrrolidine.
Compound 265
[00581] This compound was prepared via Method J using 4-(azetidin-3-ylmethoxy)-benzonitrile.
Compound 266
[00582] This compound was prepared via Method U using piperidine.
Compound 267
[00583] This compound was prepared via Method U using thiomorpholine 1 , 1 -dioxide.
Compound 268:
Figure imgf000107_0001
[00584] Cyclopropanecarboxylic acid [5-(4-ammomethyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-
2-yl]-amide (1 eq), CDI (1.1 eq) and Et3N (2.5 eq) in DCM were mixed together at 500C for Ih. The solvent was evaporated and the resulting mixture was dissolved in DMF. N-methyl-pyrazine was added to the obtained mixture. The solution was stirred at 500C for 18h. After completion of the reaction water was added and the organic phase was extracted with EtOAc. The organic layers were dried over MgSC^ and evaporated in vacuo to afford the title product purified by flash chromatography.
Compound 269
Figure imgf000108_0001
[00585] Morpholine (1 eq), CDI (1.1 eq) and Et3N (2.5 eq) in THF were mixed together at reflux for 18h. The solvent was evaporated and the resulting mixture was dissolved in acetonitrile. Methyl Iodide was added to the resulting solution. The reaction was allowed to stir at room temperature for 18h. The solvent was evaporated and the resulting mixture was dissolved in DMF. Cyclopropanecarboxylic acid [5-(4-aminomethyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide (leq) and Et3N (2.5 eq) were added to the solution which was then allowed to stirred at room temperature for 18 hrs. Water was added and the solution was extracted with EtOAc. The organic layers were dried over MgSθ4 and evaporated in vacuo to afford the title product purified by flash chromatography.
Compound 268
[00586] This compound was prepared via Method V using cyclobutanecarbonyl chloride.
Compound 269
[00587] This compound was prepared via Method B using dimethyl-piperidin-3-yl-amine.
Compound 270
[00588] This compound was prepared via Method B using piperidin-3-ol.
Compound 271
[00589] This compound was prepared via Method B using 3,3-difluoro-pyrrolidine.
Compound 272
[00590] This compound was prepared via Method V using cyclopropanecarbonyl chloride.
Compound 273 [00591] This compound was prepared via Method B using (l,l-dioxo-tetrahydrothiophen-3-yl)- methyl-amine.
Compound 274
[00592] This compound was prepared via Method B using piperidine-4-carboxylic acid amide.
Compound 275
[00593] This compound was prepared via Method B using piperidine-2-carboxylic acid amide.
Compound 276
[00594] This compound was prepared via Method B using piperidin-3-yl-methanol.
Compound 277
[00595] This compound was prepared via Method B using piperazin-2-one.
Compound 278
[00596] This compound was prepared via Method J using 4-(azetidin-3-yloxy)-benzonitrile.
Compound 279
[00597] This compound was prepared via Method J using azetidin-3-yl-carbamic acid tert-butyl ester.
Compound 280
[00598] This compound was prepared via Method J using 4-trifluoromethyl-piperidine.
Compound 281
[00599] This compound was prepared via Method J using 4-methoxy-piperidine.
Compound 282
[00600] This compound was prepared via Method J using 4-ethoxy-piperidine.
Compound 283
[00601] This compound was prepared via Method J using N-azetidin-3-yl-N-methyl-acetamide.
Compound 284
Figure imgf000110_0001
[00602] The crude cyclopropanecarboxylic acid [5-(4-bromo-phenyl)-[l,2,4]triazolo[l,5- a]pyridin-2-yl] -amide (l .O equiv.) prepared by Method A was mixed together with 4-(azetidin-3- yloxymethyl)-benzonitrile (1.2 equiv.) and potassium tert-butoxide (2.0 equiv.), in dry 1,4-dioxane (1 mL). The mixture was stirred at 800C under nitrogen. Afterwards, Pd(OAc)2 (0.1 equiv.) and BINAP (0.1 equiv.) in dry 1,4-dioxane (1 mL), were added with a syringe to the reaction mixture. The reaction was stirred overnight. LCMS showed the presence of the desired product. The mixture was filtered and submitted for preparative HPLC purification, to afford the pure product.
Compound 285
[00603] This compound was prepared via Method B using diethyl-pyrrolidin-3-yl-amine.
Compound 286
[00604] This compound was prepared via Method B using 4-phenyl-piperidin-4-ol.
Compound 287
[00605] This compound was prepared via Method B using N-azetidin-3-yl-acetamide.
Compound 288
Figure imgf000111_0001
[00606] The crude cyclopropanecarboxylic acid [5-(4-bromo-phenyl)-[l,2,4]triazolo[l,5- a]pyridin-2-yl] -amide (1.0 equiv.) prepared by method A was mixed together with azetidine-3- carbonitrile (1.2 equiv.) and potassium tert-butoxide (2.0 equiv.), in dry 1,4-dioxane (1 mL). The mixture was stirred at 800C under nitrogen. Afterwards, Pd(OAc)2 (0.1 equiv.) and BINAP (0.1 equiv.) in dry 1,4-dioxane (1 mL), were added with a syringe to the reaction mixture. The reaction was stirred overnight. LCMS showed the presence of the desired product. The mixture was filtered and submitted for preparative HPLC purification, to afford the pure product.
Compound 289
[00607] This compound was prepared via Method B using dimethyl-pyrrolidin-3-yl-amine.
Compound 290
[00608] This compound was prepared via Method B using piperidin-l-yl-piperidin-3-yl- methanone.
Compound 291
[00609] This compound was prepared via Method J using azetidin-3-yl-dimethyl-amine.
Compound 292
[00610] This compound was prepared via Method B using 3-(piperidin-4-ylmethoxy)-pyridine.
Compound 293
[00611] This compound was prepared via Method B using 4-methoxy-piperidine.
Compound 294
[00612] This compound was prepared via Method B using 4-ethoxy-piperidine Compound 295
[00613] This compound was prepared via Method B using Piperidine-3-carboxylic acid diethylamide.
Compound 296
[00614] This compound was prepared via Method B using N-piperidin-3-yl-acetamide.
Compound 297
[00615] This compound was prepared via Method P using 5-chloromethyl-pyridine-2- carbonitrile.
Compound 298
[00616] This compound was prepared via Method J using azetidin-3-ylmethyl-dimethyl-amine.
Compound 299
[00617] This compound was prepared via Method J using azetidine-3-carboxylic acid dimethylamide.
Compound 300
[00618] This compound was prepared via Method J using 4-piperidin-4-yl-morpholine.
Compound 301
[00619] This compound was prepared via Method Q using (4-hydroxy-phenyl)-acetonitrile.
Compound 302
[00620] This compound was prepared via Method J using isoxazol-3-ylamine.
Compound 303
[00621] This compound was prepared via Method J using azetidine-3-carbonitrile.
Compound 304
[00622] This compound was prepared via Method J using l,l-dioxo-tetrahydrothiophen-3- ylamine.
Compound 305
[00623] This compound was prepared via Method J using (S)-pyrrolidin-3-ol. Compound 306
[00624] This compound was prepared via Method J using 2-amino-benzamide.
Compound 307
[00625] This compound was prepared via Method using (R)-pyrrolidin-3-ol.
Compound 308
Figure imgf000113_0001
[00626] Cyclopropanecarboxylic acid [5-(4-ammomethyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-
2-yl]-amide (1 eq), CDI (1.1 eq) and Et3N (2.5 eq) in DCM were mixed together at 500C for Ih. The solvent was evaporated and the resulting mixture was dissolved in DMF. N-methyl-pyrazine was added to the obtained mixture. The solution was stirred at 500C for 18h. After completion of the reaction water was added and the organic phase was extracted with EtOAc. The organic layers were dried over MgSO4 and evaporated in vacuo to afford the title product purified by flash chromatography.
Compound 309
Figure imgf000113_0002
[00627] Morpholine (1 eq), CDI (1.1 eq) and Et3N (2.5 eq) in THF were mixed together at reflux for 18h. The solvent was evaporated and the resulting mixture was dissolved in acetonitrile. Methyl iodide was added to the resulting solution. The reaction was allowed to stir at room temperature for 18h. The solvent was evaporated and the resulting mixture was dissolved in DMF. Cyclopropanecarboxylic acid [5-(4-aminomethyl-phenyl)-[l,2,4]triazolo[l,5-a]pyridin-2-yl]-amide (leq) and Et3N (2.5 eq) were added to the solution which was then allowed to stirred at room temperature for 18 hrs. Water was added and the solution was extracted with EtOAc. The organic layers were dried over MgSθ4 and evaporated in vacuo to afford the title product purified by flash chromatography.
Compound 310
[00628] This compound was prepared via Method J using piperidin-4-yl-carbamic acid tert-butyl ester.
Compound 311
[00629] This compound was prepared via Method J using piperazin-2-one.
Compound 312
[00630] This compound was prepared via Method J using cyclopropylamine.
Compound 313
[00631] This compound was prepared via Method J using 3-hydroxy-piperidine.
Compound 314
[00632] This compound was prepared via Method J using 3,3-dimethylazetidine.
Compound 315
[00633] This compound was prepared via Method J using 3,4-difluoro-azetidine
Compound 316
[00634] This compound was prepared via Method U using pyridin-3-ylamine.
Compound 317
[00635] This compound was prepared via Method U using 3,3-difluoro-azetidine.
Compound 318
[00636] This compound was prepared via Method U using azetidine. Compound 319
[00637] This compound was prepared via Method U using 4-trifluoromethyl-piperidine.
Compound 320
[00638] This compound was prepared via Method U usinf 4,5-dimethyl piperidine.
Compound 321
[00639] This compound was prepared via Method U using 3-methoxy-azetidine.
Compound 322
[00640] This compound was prepared via Method U using N-azetidin-3-yl-acetamide.
Compound 323
[00641] This compound was prepared via Method U using N-piperidin-4-yl-acetamide.
Compound 324
[00642] This compound was prepared via Method U using azetidine-3-carboxylic acid dimethylamide.
Compound 325
[00643] This compound was prepared via Method B using 4-(azetidin-3-yloxymethyl)- benzonitrile.
Compound 326
[00644] This compound was prepared via Method B using 4-azetidin-3-yl-morpholine.
Compound 327
[00645] This compound was prepared via Method B using azetidin-3-yl-dimethyl-amine
Compound 328
[00646] This compound was prepared via Method B using azetidine-3-carbonitrile.
Compound 329
[00647] This compound was prepared via Method B using azetidin-3-ylmethyl-dimethyl-amine. Compound 330 [00648] This compound was prepared via Method B using 3,3-dimethyl-azetidine.
Compound 331
[00649] This compound was prepared via Method B using lH-[l,2,4]triazol-3-ylamine
[00650] The exemplary compounds that have been or can be prepared according to the synthetic methods described herein are listed in Table I below. The NMR spectral data of some representative compounds of the invention is given in Table II. [00651] Table I
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Table II: NMR Data of Representative Compounds of the Invention
Figure imgf000195_0002
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Biological Examples Example 1 - in vitro assays Example 1.1 JAKl inhibition assay
[00653] 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 μg 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 MgCl2, 2 μM non-radioactive ATP, 0.25 μCi 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5μL containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 μL, in a polypropylene 96-well plate (Greiner, V-bottom). After 45 min at 30 0C, reactions were stopped by adding of 25 μL/well of 150 mM phosphoric acid. All of the terminated kinase reaction was transferred to prewashed (75 mM phosphoric acid) 96 well filter plates (Perkin Elmer catalog number 6005177) using a cell harvester (Perkin Elmer). Plates were washed 6 times with 300 μL per well of a 75 mM phosphoric acid solution and the bottom of the plates was sealed. 40 μL/well of Microscint-20 was added, the top of the plates was sealed and readout was performed using the Topcount (Perkin Elmer). Kinase activity was calculated by subtracting counts per minute (cpm) obtained in the presence of a positive control inhibitor (10 μM staurosporine) from cpm obtained in the presence of vehicle. The ability of a test compound to inhibit this activity was determined as:
[00654] 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%.
[00655] Dose dilution series were prepared for the compounds enabling the testing of dose- response effects in the JAKl assay and the calculation of the IC5O for each compound. Each compound was routinely tested at concentration of 20μM followed by a 1/3 serial dilution, 8 points (20μM - 6.67μM - 2.22μM - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO. When potency of compound series increased, more dilutions were prepared and/or the top concentration were lowered (e.g. 5 μM, 1 μM). [00656] Semi-quantitative score: *> 1000nM **501-1000nM *** 101-500nM ****< 10OnM
TABLE III: JAKl IC^n Values of Compounds
Figure imgf000223_0001
Figure imgf000223_0002
Figure imgf000224_0001
Figure imgf000224_0002
Figure imgf000225_0001
Figure imgf000225_0002
Figure imgf000226_0001
Figure imgf000226_0002
Figure imgf000227_0002
Figure imgf000227_0001
Example 1.2 JΛK2 inhibition assay
[00658] 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 μg 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 μM non-radioactive ATP, 0.25 μCi 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5μL containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 μL, in a polypropylene 96-well plate (Greiner, V-bottom). After 90 min at 30 0C, reactions were stopped by adding of 25 μL/well of 150 mM phosphoric acid. All of the terminated kinase reaction was transferred to prewashed (75 mM phosphoric acid) 96 well filter plates (Perkin Elmer catalog number 6005177) using a cell harvester (Perkin Elmer). Plates were washed 6 times with 300 μL per well of a 75 mM phosphoric acid solution and the bottom of the plates was sealed. 40 μL/well of Microscint-20 was added, the top of the plates was sealed and readout was performed using the Topcount (Perkin Elmer). Kinase activity was calculated by subtracting counts per minute (cpm) obtained in the presence of a positive control inhibitor (10 μM staurosporine) from cpm obtained in the presence of vehicle. The ability of a test compound to inhibit this activity was determined as:
[00659] 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% .
[00660] Dose dilution series were prepared for the compounds enabling the testing of dose- response effects in the JAK2 assay and the calculation of the IC5O for each compound. Each compound was routinely tested at concentration of 20μM followed by a 1/3 serial dilution, 8 points (20μM - 6.67μM - 2.22μM - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO. When potency of compound series increased, more dilutions were prepared and/or the top concentration was lowered (e.g. 5 μM, 1 μM). [00661] Semi-quantitative score:
# > 100O nM
## 501-1000 nM
### 101-50O nM
#### < 100 nM
[00662] TABLE IV: JAK2 IQn Values of Compounds
Figure imgf000228_0001
Figure imgf000228_0002
Figure imgf000229_0001
Figure imgf000229_0002
Figure imgf000230_0001
Figure imgf000230_0002
Figure imgf000231_0002
Figure imgf000231_0001
Example 1.3 JAK3 inhibition assay
[00663] 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 μg 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 μM non-radioactive ATP, 0.25 μCi 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5μL containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 μL, in a polypropylene 96-well plate (Greiner, V-bottom). After 105 min at 30 0C, reactions were stopped by adding of 25 μL/well of 150 mM phosphoric acid. All of the terminated kinase reaction was transferred to prewashed (75 mM phosphoric acid) 96 well filter plates (Perkin Elmer catalog number 6005177) using a cell harvester (Perkin Elmer). Plates were washed 6 times with 300 μL per well of a 75 mM phosphoric acid solution and the bottom of the plates was sealed. 40 μL/well of Microscint-20 was added, the top of the plates was sealed and readout was performed using the Topcount (Perkin Elmer). Kinase activity was calculated by subtracting counts per minute (cpm) obtained in the presence of a positive control inhibitor (10 μM staurosporine) from cpm obtained in the presence of vehicle. The ability of atest compound to inhibit this activity was determined as:
[00664] 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%.
[00665] Dose dilution series were prepared for the compounds enabling the testing of dose- response effects in the JAK3 assay and the calculation of the IC5O for each compound. Each compound was routinely tested at concentration of 20μM followed by a 1/3 serial dilution, 8 points (20μM - 6.67μM - 2.22μM - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO. When potency of compound series increased, more dilutions were prepared and/or the top concentration was lowered (e.g. 5 μM, 1 μM). [00666] Semi-quantitative score:
+> 100O nM
++ 501-100O nM
+++ 101-50O nM
++++ < 100 nM
N/A - not available
[00667] TABLE V: JAK3 IC^n Values of Compounds
Figure imgf000232_0001
Figure imgf000232_0002
Figure imgf000233_0002
Figure imgf000233_0001
Example 1.4 TYK2 inhibition assay
[00668] 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 μg 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 μM non-radioactive ATP, 0.125 μCi 33P-gamma-ATP (GE Healthcare, catalog number AH9968) final concentrations) with or without 5μL containing test compound or vehicle (DMSO, 1% final concentration), in a total volume of 25 μL, in a polypropylene 96-well plate (Greiner, V-bottom). After 90 min at 30 0C, reactions were stopped by adding of 25 μL/well of 150 mM phosphoric acid. All of the terminated kinase reaction was transferred to prewashed (75 mM phosphoric acid) 96 well filter plates (Perkin Elmer catalog number 6005177) using a cell harvester (Perkin Elmer). Plates were washed 6 times with 300 μL per well of a 75 mM phosphoric acid solution and the bottom of the plates was sealed. 40 μL/well of Microscint-20 was added, the top of the plates was sealed and readout was performed using the Topcount (Perkin Elmer). Kinase activity was calculated by subtracting counts per minute (cpm) obtained in the presence of a positive control inhibitor (10 μM staurosporine) from cpm obtained in the presence of vehicle. The ability of a test compound to inhibit this activity was determined as:
[00669] 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%.
[00670] Dose dilution series were prepared for the compounds enabling the testing of dose- response effects in the TYK2 assay and the calculation of the IC5O for each compound. Each compound was routinely tested at concentration of 20μM followed by a 1/3 serial dilution, 8 points (20μM - 6.67μM - 2.22μM - 74OnM - 247nM - 82nM - 27nM - 9nM) in a final concentration of 1% DMSO. When potency of compound series increased, more dilutions were prepared and/or the top concentration was lowered (e.g. 5 μM, 1 μM). [00671] Semi-quantitative score:
- > 1000 nM
--501-100O nM
— 101-50O nM
— - < 10O nM
N/A - not available
[00672] TABLE VI: TYK2 ICn Values of Compounds
Figure imgf000234_0001
Figure imgf000235_0002
Figure imgf000235_0001
Example 2. Cellular assays Example 2.1 JAK-STA T signalling assay:
[00673] HeLa cells were maintained in Dulbecco's Modified Eagle's Medium (DMEM) containing 10% heat inactivated fetal calf serum, 100 U/mL penicillin and 100 μg/mL streptomycin. HeLa cells were used at 70 % confluence for transfection. 20,000 cells in 87 μL cell culture medium were transiently transfected with 40 ng pSTATl(2)-luciferase reporter (Panomics), 8 ng of LacZ reporter as internal control reporter and 52 ng of pBSK using 0.32 μL Jet-PEI (Polyp lus) as transfection reagent per well in 96-well plate format. After overnight incubation at 37°C, 10% CO2, transfection medium was removed. 75 μL of DMEM + 1.5% heat inactivated fetal calf serum was added. 15 μL of compound at 6.7x concentration was added for 60 min and then 10 μL of human OSM (Peprotech) at 33 ng/mL final concentration. [00674] All compounds were tested in duplicate starting from 20 μM followed by a 1/3 serial dilution, 8 doses in total (20 μM - 6.6 μM - 2.2 μM - 740 nM - 250 nM - 82 nM - 27 nM - 9 nM) in a final concentration of 0.2% DMSO.
[00675] After overnight incubation at 37°C, 10% CO2 cells were lysed in 100 μL lysis buffer/well (PBS, 0.9 mM CaCl2, 0.5 mM MgC12, 5% Trehalose, 0.025% Tergitol NP9, 0.15% BSA).
[00676] 40 μL of cell lysate was used to read β-galactosidase activity by adding 180 μL βGal solution (30μl ONPG 4mg/mL + 150 μL β-Galactosidase buffer (0.06 M Na2HPO4, 0.04 M NaH2PO4, 1 mM MgCl2)) for 20 min. The reaction was stopped by addition of 50 μL Na2COs 1 M. Absorbance was read at 405 nm.
[00677] Luciferase activity was measured using 40 μL cell lysate plus 40 μl of Steadylite® as described by the manufacturer (Perkin Elmer), on the Envision (Perkin Elmer).
[00678] 10 μM of a pan- JAK inhibitor was used as a positive control (100% inhibition). As negative control 0.5% DMSO (0% inhibition) was used. The positive and negative controls were used to calculate z' and 'percent inhibition' (PIN) values.
[00679] 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 %.
[00680] PIN values were plotted for compounds tested in dose-response and EC50 values were derived. [00681] TABLE VII: STAT signalling EC^Values of Compounds
* > 100O nM **501-1000 nM *** 101-50O nM **** 1- 10O nM
Figure imgf000236_0001
Figure imgf000236_0002
Figure imgf000237_0001
Figure imgf000237_0002
Figure imgf000238_0001
Figure imgf000238_0002
Figure imgf000239_0002
Figure imgf000239_0001
Example 2.2 OSM/IL-lβ signaling Assay
[00682] OSM and IL- lβ 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 μL DMEM (Invitrogen) containing 10% (v/v) FBS and 1% penicillin/streptomycin (InVitrogen) incubated at 37°C 5% CO2. Cells were preincubated with 15 μL compound in Ml 99 medium with 2% DMSO 1 hr before triggering with 15 μL OSM and IL- lβ to reach 25 ng/mL OSM and 1 ng/mL IL-I β, and MMP 13 levels were measured in conditioned medium 48 hours after triggering. MMP 13 activity was measured using an antibody capture activity assay. For this purpose, 384 well plates (NUNC, 460518, MaxiSorb black) were coated with 35 μL of a 1.5 μg/mL anti-human MMP13 antibody (R&D Systems, MAB511) solution for 24 hours at 4°C. After washing the wells 2 times with PBS + 0.05% Tween, the remaining binding sites were blocked with 100 μL 5% non-fat dry milk (Santa Cruz, sc-2325, Blotto) in PBS for 24 hours at 4°C. Next, the wells were washed 2 times with PBS + 0.05% Tween and 35 μL of 1/10 dilution of culture supernatant containing MMP13 in 100-fold diluted blocking buffer was added and incubated for 4 hours at room temperature. Next the wells were washed twice with PBS + 0.05% Tween followed by MMP13 activation by addition of 35 μL of a 1.5 mM 4- Aminophenylmercuric acetate (APMA) (Sigma, A9563) solution and incubation at 37 0C for 1 hour. The wells were washed again with PBS + 0.05% Tween and 35 μL MMP13 substrate (Biomol, P-126, OmniMMP fluorogenic substrate) was added. After incubation for 24 hours at 37°C fluorescence of the converted substrate was measured in a Perkin Elmer Wallac EnVision 2102 Multilabel Reader (wavelength excitation: 320 nm, wavelength emission: 405 nm).
[00683] 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 %.
* > 100O nM
**501-1000 nM
*** 1-50O nM
[00684] TABLE VIII: MMP13 EQn Values of Compounds
Figure imgf000240_0001
Figure imgf000240_0002
Figure imgf000241_0001
Figure imgf000241_0002
Figure imgf000242_0002
Figure imgf000242_0001
Example 2.3 PBL Proliferation assay [00685] Human peripheral blood lymphocytes (PBL) were stimulated with IL-2 and proliferation measured using a BrdU incorporation assay. The PBL were 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 were preincubated with test compounds 1 hr before IL-2 addition. Cells were cultured in RPMI 1640 containing 10% (v/v) FBS.
Example 3. In vivo models Example 3.1 CIA model
3.1.1 Materials
[00686] Completed Freund's adjuvant (CFA) and incomplete Freund's adjuvant (IFA) were purchased from Difco. Bovine collagen type II (CII), lipopolysaccharide (LPS), and Enbrel were 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.
3.1.2 Animals
[00687] Dark Agouti rats (male, 7-8 weeks old) were obtained from Harlan Laboratories
(Maison-Alfort, France). Rats 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.
3.1.3 Collagen induced arthritis (CIA)
[00688] One day before the experiment, CII solution (2 mg/mL) was prepared with 0.05 M acetic acid and stored at 4°C. Just before the immunization, equal volumes of adjuvant (IFA) and CII were mixed by a homogenizer in a pre-cooled glass bottle in an ice water bath. Extra adjuvant and prolonged homogenization might be required if an emulsion is not formed. 0.2 mL of the emulsion was injected intradermally at the base of the tail of each rat on day 1, a second booster intradermal injection (CII solution at 2 mg/mL in CFA 0.1 mL saline) was performed on day 9. This immunization method was modified from published methods (Sims NA et al., (2004) Targeting osteoclasts with zoledronic acid prevents bone destruction in collagen-induced arthritis, Arthritis Rheum. 50 2338-2346; Jou et al., 2005).
3.1.4 Study design
[00689] The therapeutic effects of the test compounds were tested in the rat CIA model. Rats were randomly divided into equal groups and each group contained 10 rats. All rats were immunized on day 1 and boosted on day 9. 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). A compound of interest was typically tested at 3 doses, e.g. 3, 10, 30 mg/kg, p.o.
3.1.5 Clinical assessment of arthritis
[00690] Arthritis was scored according the method of Khachigian 2006, Lin et al 2007 and
Nishida et al. 2004. The swelling of each of the four paws was ranked with the arthritic score as follows: 0-no symptoms; 1-mild, but definite redness and swelling of one type of joint such as the ankle or wrist, or apparent redness and swelling limited to individual digits, regardless of the number of affected digits; 2-moderate redness and swelling of two or more types of joints; 3-severe redness and swelling of the entire paw including digits; 4-maximally inflamed limb with involvement of multiple joints (maximum cumulative clinical arthritis score 16 per animal) (Nishida et al., 2004).
3.1.6 Change in body weight (%) after onset of arthritis
[00691] Clinically, body weight loss is associated with arthritis (Shelton et al., 2005; Argiles et al., 1998; Rail, 2004; Walsmith et al., 2004). Hence, changes in body weight after onset of arthritis could be used as a non-specific endpoint to evaluate the effect of therapeutics in the rat model. The change in body weight (%) after onset of arthritis was calculated as follows:
[00692]
[00693]
Figure imgf000244_0001
3.1.7 Radiology
[00694] 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-defmite 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 al, 2004; Jou et al, 2005. 3.1.8 Histology
[00695] After radiological analysis, the hind paws of mice were fixed in 10% phosphate- buffered formalin (pH 7.4), decalcified with rapid bone decalcifϊant for fine histology (Laboratories Eurobio) and embedded in paraffin. To ensure extensive evaluation of the arthritic joints, at least four serial sections (5 μm thick) were cut and each series of sections were 100 μm in between. The sections were stained with hematoxylin and eosin (H&E). Histologic examinations for synovial inflammation and bone and cartilage damage were performed double blind. In each paw, 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'.
3.1.9 Micro-computed tomography (μCT) analysis of calcaneus (heel bone):
[00696] Bone degradation observed in RA occurs especially at the cortical bone and can be revealed by μCT 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 (spaced by about 10.8 μm), are analyzed.
3.1.10 Results
[00697] The following compounds were efficacious in all readouts performed in the rat CIA study, with statistical significance in several of the readouts: 18, 37, 145, 176, 200, 215, and 330.
Example 3.2 Septic shock model
[00698] Injection of lipopolysaccharide (LPS) induces a rapid release of soluble tumour necrosis factor (TNF-alpha) into the periphery. This model is used to analyse prospective blockers of TNF release in vivo.
[00699] Six BALB/cJ female mice (20 g) per group were treated at the intended dosing once, po.
Thirty minutes later, LPS (15 μg/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 μg/kg) was used as a reference antiinflammatory compound. Selected compounds are tested at one or multiple doses, e.g. 3 and/or 10 and/or 30 mg/kg, po. [00700] The following compounds exhibited a statistically significant reduction in the TNF release (>50%) at 30mg/kg po: 12, 18, 36, 37, 52, 60, 74, 125, 148, 176, 197, 200, 207, 208, 215, and 229.
Example 3.3 MAB model
[00701] 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 were injected i.v. with a cocktail of mAbs directed against collagen II. One day later, compound treatment was initiated (vehicle: 10% (v/v) HPβCD). Three days later, mice received an i.p. LPS injection (50 μg/mouse), resulting in a fast onset of inflammation. Compound treatment was continued until 10 days after the mAb injection. Inflammation was read by measuring paw swelling and recording the clinical score of each paw. The cumulative clinical arthritis score of four limbs was 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.
0 Symptom free
1 Mild, but definite redness and swelling of one type of joint such as the ankle or wrist, or apparent redness and swelling limited to individual digits, regardless of the number of affected digits
2 Moderate redness and swelling of two or more types of joints
3 Severe redness and swelling of the entire paw including digits
4 Maximally inflamed limb with involvement of multiple joints
[00702] The following compound, dosed p.o. at 30 mg/kg reduced the clinical score with statistical significance at 30 mg/kg and significantly reduced inflammation at 30 mg/kg doses: 36, 37, 176
Example 3.4 Oncology models
[00703] In vitro and in vivo models to validate efficacy of small molecules towards JAK2-driven myleoproliferative diseases are described by Wernig et al. Cancer Cell 13, 311, 2008 and Geron et al. Cancer Cell 13, 321, 2008.
Example 3.5 Mouse IBD model
[00704] In vitro and in vivo models to validate efficacy of small molecules towards IBD are described by Wirtz et al. 2007.
Example 3.6 Mouse Asthma model [00705] In vitro and in vivo models to validate efficacy of small molecules towards asthma are described by Nials et al, 2008; Ip et al. 2006; Pernis et al, 2002; Kudlacz et al, 2008)
Example 4: Toxicity, DMPK and Safety Models
Example 4.1 Thermodynamic solubility
[00706] A solution of 1 mg/mL of the test compound is prepared in a 0.2M phosphate buffer pH7.4 or a 0.1M citrate buffer pH3.0 at room temperature in a glass vial.
[00707] The samples are rotated in a Rotator drive STR 4 (Stuart Scientific, Bibby) at speed 3.0 at room temperature for 24 hours.
[00708] After 24 hours, 800μL of the sample is transferred to an eppendorf tube and centrifuged
5 min at 14000rpm. 200 μL of the supernatant of the sample is then transferred to a MultiscreenR
Solubility Plate (Millipore, MSSLBPC50) and the supernatant is filtered (10-12" Hg) with the aid of a vacuum manifold into a clean Greiner polypropylene V-bottom 96well plate (Cat no.651201). 5 μL of the filtrate is diluted into 95 μL (F20) of the same buffer used to incubate in the plate containing the standard curve (Greiner, Cat no. 651201).
[00709] The standard curve for the compound is prepared freshly in DMSO starting from a
1OmM DMSO stock solution diluted factor 2 in DMSO (5000μM) and then further diluted in DMSO up to 19.5μM. 3μl of the dilution series as from 5000μM is then transferred to a 97μl acetonitrile-buffer mixture (50/50). The final concentration range was 2.5 to 150 μM.
[00710] 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
Quanoptimize to determine the appropriate mass of the molecule.
[00711] The samples are analyzed on LCMS with a flow rate of lml/min. Solvent A is 15mM ammonia and solvent B is acetonitrile. The sample is run under positive ion spray on an XBridge Cl 8
3.5μM (2.1 x 30mm) column, from Waters. The solvent gradient has a total run time of 2 minutes and ranges from 5% B to 95% B.
[00712] Peak areas are analyzed with the aid of Masslynx software package and peak areas of the samples are plotted against the standard curve to obtain the solubility of the compound.
[00713] Solubility values are reported in μM or μg/mL.
Example 4.2 Aqueous Solubility
[00714] Starting from a 1OmM stock in DMSO, a serial dilution of the compound is prepared in
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. [00715] The final concentration ranges from 200μM to 2.5μM in 5 equal dilution steps. The final DMSO concentration does not exceed 2%. 200μM 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.
[00716] 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 was plotted onto a graph. The first concentration at which the compound appears completely dissolved is the concentration is reported, however the true concentration lies somewhere between this concentration and one dilution step higher.
[00717] Solubility values are reported in μg/mL.
Example 4.3 Plasma Protein Binding (Equilibrium Dialysis)
[00718] 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 lOμM and final DMSO concentration of 0.5% (5.5μl in 1094.5μl plasma in a PP-Masterblock 96well (Greiner, Cat no. 780285))
[00719] A Pierce Red Device plate with inserts (ThermoScientific, Cat no. 89809) is prepared and filled with 750μL PBS in the buffer chamber and 500μL of the spiked plasma in the plasma chamber. The plate is incubated for 4 hours at 37°C while shaking at 230rpm. After incubation, 120μL of both chambers is transferred to 360μL 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, 651201) for analysis on LCMS.
[00720] 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 Quanoptimize to determine the appropriate mass of the molecule.
[00721] The samples are analyzed on LCMS with a flow rate of lmL/min. Solvent A was 15mM ammonia and solvent B was acetonitrile. The sample was run under positive ion spray on an XBridge
Cl 8 3.5μM (2.1 x 30mm) column, from Waters. The solvent gradient has a total run time of 2 minutes and ranges from 5% B to 95% B.
[00722] 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 is reported to the LIMS as percentage bound to plasma.
[00723] The solubility of the compound in the final test concentration in PBS is inspected by microscope to indicate whether precipitation is observed or not. Example 4.4 Liability for QT prolongation [00724] Potential for QT prolongation is assessed in the hERG patch clamp assay.
4.4.1 Conventional whole-cell patch-clamp
[00725] Whole-cell patch-clamp recordings are performed using an EPClO amplifier controlled by Pulse v8.77 software (HEKA). Series resistance is typically less than 10 MΩ and compensated by greater than 60%, recordings are not leak subtracted. Electrodes are manufactured from GC 150TF pipette glass (Harvard), resistance is between 2 and 3 MΩ.
[00726] The external bathing solution contains: 135 mM NaCl, 5 mM KCl, 1.8 mM CaCl2, 5 mM Glucose, 10 mM HEPES, pH 7.4.
[00727] The internal patch pipette solution contains: 10OmM Kgluconate, 20 mM KCl, ImM
CaCl2, 1 mM MgCl2, 5mM Na2ATP, 2mM Glutathione, 11 mM EGTA, 10 mM HEPES, pH 7.2.
[00728] Drugs are perfused using a Biologic MEV-9/EVH-9 rapid perfusion system.
[00729] All recordings are performed on HEK293 cells stably expressing hERG channels. Cells are cultured on 12 mm round coverslips (German glass, Bellco) anchored in the recording chamber using two platinum rods (Goodfellow). hERG currents are evoked using an activating pulse to +40 mV for 1000 ms followed by a tail current pulse to -50 mV for 2000 ms, holding potential was -80 mV.
Pulses are applied every 20s and all experiments are performed at room temperature.
4.4.2 Data Analysis
[00730] IC5O and IC2O values are calculated for each compound tested. The fold difference between the IC2O and the unbound Cmax concentrations of the test compound obtained at relevant therapeutic doses as determined by results obtained from the rat CIA model is calculated.
[00731] For the concentration response curves, peak tail current amplitude is measured during the voltage step to -50 mV. Curve- fitting of concentration-response data is performed using the equation: y = a + [( b -a )/ ( 1+ 10Λ ( ( logc-x ) d )]
[00732] where a is minimum response, b is maximum response and d is Hill slope, this equation can be used to calculate both IC5O (where y = 50 and c is the IC5O value) and IC2O (where y = 20 and c is the IC2O value). GraphPad® Prism® (Graphpad® Software Inc.) software is used for all curve fitting. [00733] A difference of 100 fold or greater indicates a low potential for QT prolongation.
Example 4.5 Microsomal stability
[00734] A I OmM stock solution of compound in DMSO was diluted 1000 fold in a 1 S2 mM phosphate buffer pH7/l iυ a % deep well plate (Greiner, Cat no,7802ϊJ5j and prc-incubated at 37°C. [00735] 40μL of deionised water was added to a well of a polypropylene Matrix 2D barcode labelled storage tube (Thermo Scientific) and pre-ineubated at 37°C.
[00736] A Glucose-6-phophate-dehydrogenase (GόPDH) working stock solution was prepared in 182mM. phosphate buffer pH7,4 and placed on ice before use. A co-factor containing MgC12, gIucose-6-phosphate and N ADP+ was prepared in deionised water and placed on ice before use.
[00737] A final working solution containing liver microsomes (Xenotech) of a species of interest
(human, mouse, rat, dog), previously described G6PDH and co- factors was prepared and this mix was incubated for no longer than 20 minutes at. room temperature.
[00738] 30μL of the pre-heated compound dilution was added to 40μL of pre-heated water in the Matrix tubes and 30μL of the microsomal mix was added. Final reaction concentrations were 3μM compound, lmg microsomes, 0.4LVmL GDPDH, 3.3mM MgCl?., 3.3mM glucose-6-phosphate and
1.3mM NADP+.
[00739] To measure percentage remaining of compound at time zero MeOH or ACN was added
(1:1) to the well before adding the microsomal mix. The plates were sealed with Matrix Sepra seaisTM
(Matrix, Cat No.4464) and shaken for a few seconds ensure complete mixing of all components.
[00740] The samples which were not slopped are incubated at 37°C, 300rpm and after 1 hour of incubation the reaction was stopped with MeOH or ACN (1:1).
[00741] After stopping the reaction the samples were mixed and placed on ice for 30min to precipitate the proteins. The plates were then centrifuged 30 mm at 1200ref at 4°C and the supernatant was transferred to a 96 v-bottom PP plate (Greiner, 651201) for analysis on LCMS.
[00742] These plates were sealed with sealing mats (MA96RD-04S) of www.kinesis.co.uk and samples were measured at room temperature on LCMS (ZQ 1525 from Waters) under optimized conditions using Quanoptimize to determine the appropriate mass of the parent molecule.
[00743] The samples were analyzed on LCMS with a flow rate of imL/min. Solvent A was
15mM ammonia and solvent B was methanol or acetonitrile, depending on the stop solution used. The samples were run under positive ion spray on an XBridge Cl 8 3.5μM (2.1 x 30mm) column, from
Waters. The solvent gradient had a total run time of 2 minutes and ranges from 5% B to 95% B.
Peak area from the parent compound at time 0 was considered to be 100% remaining. The percentage remaining after 1 hour incubation was calculated from time 0 and was calculated as the percentage remaining. The solubility of the compound in the final test concentration in buffer is inspected by microscope and results are reported.
[00744] The data on microsomal stability are expressed as a percentage of the total amount of compound remaining after 60 minutes.
* 0-25
** 26-50
*** 51-75 **** 76-100 ABLE IX: Microsomal Stability of Compounds
Figure imgf000251_0001
Figure imgf000252_0001
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Example 4.6 Caco2 Permeability
[00746] Bi-directional 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).
[00747] 2x105 cells/well were seeded in plating medium consisting of DMEM + GlutaMAXI +
1% NEAA + 10% FBS (FetalClone II) + 1% Pen/Strep. The medium was changed every 2 - 3 days.
[00748] 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 (125μL) or basolateral (600μL) chambers of the Transwell plate assembly at a concentration of 10 μM with a final DMSO concentration of 0.25%.
[00749] 50μM 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.
[00750] After a 1 hour incubation at 37°C while shaking at an orbital shaker at 150rpm, 70μL aliquots were taken from both apical (A) and basal (B) chambers and added to lOOμLl 50:50 acetonitrile: water solution containing analytical internal standard (0.5μM carbamazepine) in a 96 well plate.
[00751] Lucifer yellow was measured with a Spectramax Gemini XS (Ex 426nm and Em
538nm) in a clean 96 well plate containing 150μL of liquid from basolateral and apical side.
[00752] Concentrations of compound in the samples were measured by high performance liquid- chromatography/mass spectroscopy (LC-MS/MS).
[00753] Apparent permeability (Papp) values were calculated from the relationship: Papp = [COmpOUnd] acceptor final X VaCceptor / ([COmpOUnd] donor initial X Vdonor) / Tmc X Vdonor / Surface area X 60 X
10"6 cm/s
V = chamber volume
Tmc = incubation time.
Surface area = 0.33cm
[00754] The Efflux ratios, as an indication of active efflux from the apical cell surface, were calculated using the ratio of Papp B>A/ Papp A>B.
[00755] The following assay acceptance criteria were used:
Propranolol: Papp (A>B) value > 20(χl0"6 cm/s)
Rhodamine 123 or Vinblastine: Papp (A>B) value < 5 (xlO 6 cm/s) with Efflux ratio >5.
Lucifer yellow permeability: <100 nm/s
[00756] TABLE X: Papp and Efflux Values of Compounds
Figure imgf000256_0001
Figure imgf000256_0002
Figure imgf000257_0001
Figure imgf000257_0002
Example 4.7 Pharmacokinetic study in rodents
3.1.3 Pharmacokinetic study
[00757] 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-β-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 -200C pending analysis.
3.1.4 Quantification of compound levels in plasma
[00758] Plasma concentrations of each test compound are determined by an LC-MS/MS method in which the mass spectrometer was operated in positive electrospray mode.
3.7.5 Determination of pharmacokinetic parameters [00759] Pharmacokinetic parameters are calculated using Winnonlin® (Pharsight®, United
States). Example 4.8 7-Day rat toxicity study
[00760] A 7-day oral toxicity study with test compounds is 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.
[00761] The test compounds are formulated in 30% (v/v) HPβCD in purified water. Each group included 5 principal male rats as well as 3 satellite animals for toxicokinetics. A fourth group is given 30% (v/v) HPβCD in water only, at the same frequency, dosage volume and by the same route of administration, and acted as the vehicle control group.
[00762] The goal of the study is to determine the lowest dose that resulted in no adverse events being identified (no observable adverse effect level - NOAEL).
[00763] It will be appreciated by those skilled in the art that the foregoing descriptions are exemplary and explanatory in nature, an as indiced intended to illustrate the invention and its preferred embodiments. Through routine experimentation, an artisan will recognise apparent modifications and variations that may be made without departing from the spirit of the invention. Thus, the invention is intended to be defined not by the above description, but by the following claims and their equivalents.
[00764] REFERENCES
Choy EH, Panayi GS. (2001). N Engl J Med. 344: 907-16.
Chubinskaya S and Kuettner KE (2003). Regulation of osteogenic proteins by chondrocytes. The international journal of biochemistry & cell biology 35(9)1323-1340. Clegg DO et al. (2006) N Engl J Med. 2006 354:795-808. Glucosamine, chondroitin sulfate, and the two in combination for painful knee osteoarthritis. Firestein GS. (2003). Nature. 423:356-61.
Kachigian LM. (2006) Collagen antibody-induced arthritis, Nature Protocols 2512-2516: Lee DM, Weinblatt ME (2001). Lancet. 358: 903-11. Legendre F, Dudhia J, Pujol J-P, Bogdanowicz P. (2003) JAK/STAT but not ERK1/ERK2 pathway mediates interleuking (IL)-6/soluble IL-6R down-regulation of type II collagen, aggrecan core, and link protein transcription in articular chondrocytes. J Biol Chem. 278(5)2903-2912. Li WQ, Dehnade F, Zafarullah M. (2001) Oncostatin M-induced matrix metalloproteinase and tissue inhibitor of metalloproteinase-3 genes expression in chondrocytes requires janus kinase/STAT signaling pathway. (2001) J Immunol 166:3491-3498.
O'Dell JR. (2004) Therapeutic strategies for rheumatoid arthritis. N Engl J Med. 350(25):2591-602. Osaki M, Tan L, Choy BK, Yoshida Y, Cheah KSE, Auron PE, Goldring MB. (2003) The TATA- conatining core promoter of the type II collagen gene (COL2A1) is the target of interferon- gamma-mediated inhibition in human chondrocytes: requirement for STATl alpha, JAKl and
JAK2. Biochem J 369:103-115. Oste L et al., ECTC Montreal 2007: A high throughput method of measuring bone architectural disturbance in a murine CIA model by micro-CT morphometry Otero M, Lago R, Lago F, Gomez Reino JJ, Gualillo O. (2005) Signalling pathway involved in nitric oxide synthase type II activation in chondrocytes: synergistic effect of leptin with interleukin-1.
Arthritis Research & Therapy 7:R581-R591. Rodig SJ, Meraz MA, White JM, Lampe PA, Riley JK, Arthur CD, King KL, Sheehan KCF, Yin L,
Pennica D, Johnson EM, Schreiber RD. (1998) Disruption of the Jakl gene demonstrates obligatory and nonredundant roles of the jaks in cytokine-induced biologic responses Cell 93:
373-383.
Sims NA et al., (2004) Targeting osteoclasts with zoledronic acid prevents bone destruction in collagen- induced arthritis, Arthritis Rheum. 50 2338-2346: Smolen JS, Steiner G. (2003). Nat Rev Drug Discov. 2: 473-88. Wernig et al. (2008) Efficacy of TGl 01348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera, Cancer Cell 13(4), 311-320 Geron et al. (2008) Selective inhibition of JAK2-driven erythroid differentiation of polycythemia vera progenitors Cancer Cell 13 (4), 321-30 Wieland HA, Michaelis M, Kirschbaum BJ, Rudolphi KA. (2005). Nat Rev Drug Discov. 4:331-44.
Osteoarthritis - an unbeatable disease? Wirtz et al. (2007) Mouse Models of Inflammatory Bowel Disease, Advanced Drug Delivery Reviews,
2007, 1073-1083: Tarn, L., McGlynn, L.M., Traynor, P., Mukherjee, R., Bartlett, J.M.S., Edwards, J. (2007) British
Journal of Cancer, 97, 378-383
Constantinescu et al., 2007, Trends in Biochemical Sciences 33(3): 122-131
Tetsuji Naka, Norihiro Nishimoto and Tadamitsu Kishimoto, Arthritis Res 2002, 4 (suppl 3):S233-S242 O'Shea, J.J., Pesu, M., Borie, D.C., Changelian, P.S., Nature Reviews, 2004, 555-564 Nials et al. (2008) Mouse Models of Allergic Asthma: Acute and Chronic Allergen Challenge, Disease
Models & Mechanisms, 213-220. Ip et al. (2006) 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. Pernis et al. (2002) JAK-STAT signaling in asthma J. Clin. Invest. 1279. Kudlacz et al. (2008) 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, Su X, Liu W, Devidas M, Atlas SR, Chen I-M, Clifford RJ, Gerhard DS, Carroll WL, Reaman GH, Smith M, Downing JR, Hunger SP Willmane CL; (2009) JAK mutations in high- risk childhood acute lymphoblastic leukemia, PNAS May 22. [Epub ahead of print] Argiles JM, Lopez-Soriano FJ. (1998)Catabolic proinflammatory cytokines. Curr Opin Clin Nutr Metab
Care. 1 :245-51. Bush KA, Farmer KM, Walker JS, Kirkham BW. (2002) Reduction of joint inflammation and bone erosion in rat adjuvant arthritis by treatment with interleukin-17 receptor IgGl Fc fusion protein.
Arthritis Rheum. 46: 802-5. Jou IM, Shiau AL, Chen SY, Wang CR, Shieh DB, Tsai CS, Wu CL. (2005) Thrombospondin 1 as an effective gene therapeutic strategy in collagen-induced arthritis. Arthritis Rheum. 52:339-44. Nishida K, Komiyama T, Miyazawa S, Shen ZN, Furumatsu T, Doi H, Yoshida A, Yamana J,
Yamamura M, Ninomiya Y, Inoue H, Asahara H. (2004) Histone deacetylase inhibitor suppression of autoantibody- mediated arthritis in mice via regulation of pl6INK4a and p21(WAFl/Cipl) expression. Arthritis Rheum. 10: 3365-76. Rail LC, Roubenoff R. (2004) Rheumatoid cachexia: metabolic abnormalities, mechanisms and interventions. Rheumatology; 10:1219-23. Salvemini D, Mazzon E, Dugo L, Serraino I, De Sarro A, Caputi AP, Cuzzocrea S. (2001) Amelioration of joint disease in a rat model of collagen- induced arthritis by M40403, a superoxide dismutase mimetic. Arthritis Rheum. 44:2909-21. Shelton DL, Zeller J, Ho WH, Pons J, Rosenthal A. (2005) Nerve growth factor mediates hyperalgesia and cachexia in auto-immune arthritis. Pain. 116:8-16. Sims NA, Green JR, Glatt M, Schlict S, Martin TJ, Gillespie MT, Romas E. (2004) Targeting osteoclasts with zoledronic acid prevents bone destruction in collagen-induced arthritis. Arthritis
Rheum., 50: 2338-46. Walsmith J, Abad L, Kehayias J, Roubenoff R. (2004) Tumor necrosis factor-alpha production is associated with less body cell mass in women with rheumatoid arthritis. J Rheumatol.; 31 :23-9. Khachigian, L. M. Collagen antibody- induced arthritis. (2006) Nature Protocols 1, 2512-6. Lin HS, Hu CY, Chan HY, Liew YY, Huang HP, Lepescheux L, Bastianelli E, Baron R, Rawadi G,
Clement-Lacroix P. (2007) Anti-rheumatic activities of histone deacetylase (HDAC) inhibitors in vivo in collagen-induced arthritis in rodents. Br J Pharmacol. Apr;150 (7):829-31.
[00765] All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth. [00766] From the foregoing description, various modifications and changes in the compositions and methods of this invention will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein. [00767] It should be understood that factors such as the differential cell penetration capacity of the various compounds can contribute to discrepancies between the activity of the compounds in the in vitro biochemical and cellular assays.
[00768] At least some of the chemical names of compounds of the invention as given and set forth in this application, may have been generated on an automated basis by use of a commercially available chemical naming software program, and have not been independently verified. Representative programs performing this function include the Lexichem naming tool sold by Open Eye Software, Inc. and the Autonom Software tool sold by MDL, Inc. In the instance where the indicated chemical name and the depicted structure differ, the depicted structure will control.
[00769] Chemical structures shown herein were prepared using either ChemDraw® or ISIS®
/DRAW. Any open valency appearing on a carbon, oxygen or nitrogen atom in the structures herein indicates the presence of a hydrogen atom. Where a chiral center exists in a structure but no specific stereochemistry is shown for the chiral center, both enantiomers associated with the chiral structure are encompassed by the structure.

Claims

WHAT IS CLAIMED IS:
1. A compound according to Formula I:
Figure imgf000262_0001
wherein
CyI is selected from aryl and heteroaryl;
Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)- , -S(O)2-, - N(R4a)CO-, -CH2-N(R4a)- or - N(R4a)SO2-; each R1 is independently selected from CpC6 alkyl, substituted CpC6 alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted Ci-Ce alkoxy, substituted or unsubstituted amido, substituted or unsubstituted amino, substituted sulfmyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from Ci-Cβ alkyl, substituted Ci-Ce alkyl, acyl, substituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted CpC6 alkoxy, substituted or unsubstituted amido, alkoxycarbonyl, substituted alkoxycarbonyl, arylalkyloxy, substituted arylalkyloxy, substituted or unsubstituted amino, aryl, substituted aryl, arylalkyl, substituted sulfanyl, substituted sulfmyl, substituted sulfonyl, substituted or unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, halo, substituted or unsubstituted heteroaryl, hydroxyl, nitro, and thiol;
R a is selected from substituted or unsubstituted Ci-Cβ alkyl and substituted or unsubstituted C3- C7 cycloalkyl;
R3b is independently selected from substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5-10 membered heteroaryl; or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from substituted Ci-Cβ alkyl, substituted or unsubstituted aryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl, substituted or unsubstituted 5- 10 membered heteroaryl; each R4a, R4b and R4c is independently selected from H, Ci-C6 alkyl, substituted Ci-C6 alkyl, C3- C7 cycloalkyl, or substituted C3-C7 cycloalkyl, substituted or unsubstituted acyl; ml is 0, 1, or 2; m2 is 0, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that i) when Ll is -O-, -N(R4a)-, -CH2-N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1, 2, 3, or 4; ii) when CyI is Ph, Ll is a bond, nl is 0, and R3b is -OR3c, then R3c is other than Me or CF3; or a pharmaceutically acceptable salt thereof, in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases. 2. The compound according to claim 1, wherein
CyI is selected from aryl and heteroaryl;
Ll is selected from a single bond, -O-, -C(O)-, -C[=N(R4a)]-, -N(R4a)-, -CON(R4a)-, -SO2N(R4a)- , -S(O)2-, - N(R4a)CO-, -CH2-N(R4a)-or - N(R4a)SO2-; each R1 is independently selected from unsubstituted CpC6 alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted Ci-Ce alkoxy, unsubstituted amido, unsubstituted amino, unsubstituted aminosulfonyl, sulfonic acid, sulfonic acid ester, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, and hydroxyl; each R a is independently selected from unsubstituted Ci-Cβ alkyl, unsubstituted acyl, unsubstituted acylamino, unsubstituted CpC6 alkoxy, unsubstituted amido, unsubstituted alkoxycarbonyl, unsubstituted arylalkyloxy, unsubstituted amino, unsubstituted aryl, unsubstituted arylalkyl, aminosulfonyl (which aminosulfonyl may be substituted with unsubstituted CpC4 alkyl), sulfonic acid, sulfonic acid ester, azido, carboxy, cyano, unsubstituted C3-C7 cycloalkyl, unsubstituted 4-7 membered heterocycloalkyl, halo, unsubstituted heteroaryl, hydroxyl, nitro, and thiol;
R a is selected from unsubstituted Ci-Ce alkyl and unsubstituted C3-C7 cycloalkyl;
R3b is independently selected from aryl (which aryl may be substituted with halo, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted C1-C4 alkyl, unsubstituted C1-C4 alkoxy, unsubstituted d-C4 haloalkyl, unsubstituted Ci-C4haloalkoxy, unsubstituted 5-7-membered heteroaryl, unsubstituted acylamino, unsubstituted amino, cyano, -(CH2) I-4-CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted with cyano), 4-7 membered heterocycloalkyl (which 4-7 membered heterocycloalkyl may be substituted with C1-C4 alkyl (which C1-C4 alkyl may be substituted with aryl, heteroaryl, heterocycloalkyl, C1-C4 alkoxy, -O-aryl, -0-heteroaryl, OH), unsubstituted Ci-C4haloalkyl, aryl (which aryl may be substituted with halo, unsubstituted C1-C4 alkoxy), OH, halo, cyano, acyl (which acyl may be substituted with unsubstituted aryl, unsubstituted heterocycloalkyl, unsubstituted CpC4 alkyl), heteroaryl (which heteroaryl may be substituted with halo), CpC4 dialkylamino, unsubstituted 4-7 membered heterocycloalkyl, unsubstituted -O -heteroaryl, amido (which heteroaryl may be substituted with unsubstituted C1-C4 alkyl), unsubstituted C1-C4 alkoxy), 5-10 membered heteroaryl (which heteroaryl may be substituted with C1-C4 alkyl (which Ci-C4 alkyl may be substituted with unsubstituted aryl), unsubstituted CpC4 haloalkyl, unsubstituted amido, halo, 4-7-membered heterocycloalkyl (which heterocycloalkyl may be substituted with unsubstituted CpC4 alkyl), unsubstituted CpC4 alkoxy, CN, unsubstituted C3-C7 cycloalkyl, OH, aryl (which aryl may be substituted with unsubstituted CpC4 haloalkyl) unsubstituted 5-7-membered heteroaryl, carboxy (which carboxy may be substituted with unsubstituted CpC4 alkyl)), or R3b is independently selected from O-R3c, NH- R3c, CO-R3c, and CON(R4a)-R3c; and R3c is independently selected from
Ci-Ce alkyl (which CpC4 alkyl is substituted with aryl (which aryl may be substituted with halo, CN, unsubstituted CpC4 alkyl, unsubstituted 5-7-membered heterocycloalkyl, unsubstituted 5-10 membered heteroaryl), 5-10 membered heteroaryl (which heteroaryl may be substituted with unsubstituted CpC4 alkyl)), aryl (which aryl may be substituted with halo, CN, unsubstituted CpC4 alkoxy, unsubstituted amido, unsubstituted CpC4 alkyl, unsubstituted CpC4, haloalkyl, -(CH2) 1-4-CN), C3-C7 cycloalkyl (which C3-C7 cycloalkyl may be substituted withunsubstituted 4-7 membered heterocycloalkyl, 5-10 membered heteroaryl (which heteroaryl may be substituted with halo, cyano, unsubstituted CpC4 alkyl, unsubstituted CpC4 alkoxy, unsubstituted 4-7 membered heterocycloalkyl, ); each R4a, R4b and R4c is independently selected from H, CpC6 alkyl (which CpC4 alkyl may be substituted with unsubstituted CpC4 alkoxy, unsubstituted dialkylamino, unsubstituted 4-7 membered heterocycloalkyl), unsubstituted C3-C7 cycloalkyl, acyl (which acyl may be substituted with unsubstituted CpC6 alkyl); ml is 0, 1, or 2; m2 is 0, 1, 2, or 3; and nl is 0, 1, 2, 3, or 4; provided that when Ll is -O-, -N(R4a)-, -CON(R4a)-, or -SO2N(R4a)-, and R3b is other than cycloalkyl, aryl or 5-10 membered heteroaryl, then nl is 1,
2, 3, or 4; or a pharmaceutically acceptable salt thereof, in the treatment and./or prevention 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. diseases involving the anabolic stimulation of chondrocytes), congenital cartilage malformations, diseases associated with hypersecretion of IL6 and transplantation rejection (e.g. organ transplant rejection) or proliferative diseases.
3. The compound, or pharmaceutically acceptable salt thereof, according to claim 1 or 2, wherein ml is 0.
4. The compound, or pharmaceutically acceptable salt thereof, according to claim 1 or 2 wherein ml is 1 or 2; and each R1 is independently selected from Ci-Cβalkyl, substituted Ci-Cβalkyl, and halo.
5. The compound, or pharmaceutically acceptable salt thereof, according to claim 4 wherein ml is 1 or 2; and each R1 is independently selected from Me, CF3, Cl and F.
6. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1 to 5 wherein R a is substituted or unsubstituted Ci-Ce alkyl.
7. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1 to 5 wherein R a is substituted or unsubstituted C3-C7 cycloalkyl.
8. The compound, or pharmaceutically acceptable salt thereof, according to claim 7 wherein R2a is cyclopropyl, cyclobutyl, or cyclopentyl.
9. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1 to 8 wherein R4b and R4c are independently selected from H and Me.
10. The compound, or pharmaceutically acceptable salt thereof, according to claim 1 or 2 wherein the compound is according to Formula II:
Figure imgf000265_0001
wherein CyI, Ll, R3a, R3b, m2, and nl are as in claim 1 or 2.
11. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1-
10, wherein CyI is phenyl; and m2 is 0.
12. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1- 10, wherein CyI is phenyl; m2 is 1, 2 or 3; and each R3a is independently CpC6 alkyl, CpC6 haloalkyl, Ci-Ce alkoxy, or halo.
13. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1- 10, wherein CyI is phenyl; m2 is 1, 2 or 3; and each R3a is independently Cl, F, Me, Et, OMe, CF3, CONH2, CONMe2, CONHMe, CN, NHCOMe, COOH, OH or COOEt.
14. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1- 10, wherein CyI is 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 benzodioxanyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted quinolinyl, or substituted or unsubstituted isoquinolinyl; and m2 is θ.
15. The compound, or pharmaceutically acceptable salt thereof, according to claim 1 or 2 wherein the compound is according to Formula III:
Figure imgf000266_0001
wherein Ll, R3b, and nl are as in claim 1 or 2.
16. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1-15 wherein R3b is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted C3-C7 cycloalkyl, or substituted or unsubstituted 4-7 membered heterocycloalkyl.
17. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 1-15 wherein Ll is selected from a single bond, -0-, -N(R4a)-, -C(O)-, -C[=N(R4a)]-, -C0N(R4a)-, - SO2N(R4a)-, -S(O)2-, -N(R4a)SO2- and -N(R4a)C0-; nl is O, 1, 2, 3, or 4; and R3b is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted C3-C7 cycloalkyl, substituted or unsubstituted 4-7 membered heterocycloalkyl.
18. The compound, or pharmaceutically acceptable salt thereof, according to claim 17 wherein R3b is substituted or unsubstituted C3-C7 cycloalkyl.
19. The compound, or pharmaceutically acceptable salt thereof, according to claim 18 wherein R3b is substituted or unsubstituted cyclopropyl, substituted or unsubstituted cyclobutyl, substituted or unsubstituted cyclohexyl, or substituted or unsubstituted cyclopentyl.
20. The compound, or pharmaceutically acceptable salt thereof, according to claim 17 wherein R3b is substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.
21. The compound, or pharmaceutically acceptable salt thereof, according to claim 20 wherein R3b 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 tetrazolyl, substituted or unsubstituted oxazolyl, substituted or unsubstituted oxadiazolyl, substituted or unsubstituted thiazolyl, substituted or unsubstituted thiophenyl, substituted or unsubstituted indolyl, substituted or unsubstituted indazolyl, substituted or unsubstituted benzimidazolyl, substituted or unsubstituted benzofuranyl, substituted or unsubstituted benzodioxanyl, substituted or unsubstituted benzoxazolyl, substituted or unsubstituted quinolinyl, or substituted or unsubstituted isoquinolinyl.
22. The compound, or pharmaceutically acceptable salt thereof, according to claims 17 wherein R3b is substituted or unsubstituted 4-7-membered heterocycloalkyl, provided that: when the heterocycle is attached via an heteroatom, and Ll is -O-, -N(R4a)-, -SO2N(R4a)-, and - CON(R4a)-, nl is not 0 or 1.
23. The compound, or pharmaceutically acceptable salt thereof, according to claim 22 wherein R3b is piperidinyl, morpholinyl, piperazinyl, homopiperazinyl or pyrrolidinyl, each of which may be unsubstituted or substituted with Ci-Cβalkyl, acyl, phenyl, or OH, provided that: when the heterocycle is attached via an heteroatom, and Ll is -O-, -N(R4a)-, -SO2N(R421)-, and - CON(R4a)-, nl is not 0 or 1.
24. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 15- 23, wherein R4a is H, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C1-C4 alkoxy.
25. The compound, or pharmaceutically acceptable salt thereof, according to claim 24, wherein R4a is H.
26. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 15- 23, wherein R4a is -(CH2)n2-R6a; wherein n2 is 0, 1, or 2; and R6a is H, CN, NMe2, or tetrahydrofuranyl.
27. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 15- 23, wherein R4a is -CH(CH3)-(CH2)n2-R6a; wherein n2 is 0 or 1 and R6a is H, or OMe.
28. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 15- 23, wherein R3b is OPh, or 0-(4-F-Ph).
29. The compound, or pharmaceutically acceptable salt thereof, according to any one of claims 15- 23, wherein R3b is CO-R3c and R3c is:
Figure imgf000267_0001
30. The compound, or pharmaceutically acceptable salt thereof, according to claim 15, wherein the
3b .
-Ph-Ll -(CH2)ni-R is selected from:
Figure imgf000268_0001
wherein n2 is nl; R3b, and n2 are as in claim 1 or 2; and Cy3 is substituted or unsubstituted 4-7 membered N containing 4-7-membered heterocycloalkyl.
31. The compound, or pharmaceutically acceptable salt thereof, according to claim 15 or 30 wherein R3b is selected from:
Figure imgf000268_0002
Figure imgf000269_0001
wherein each R5a is independently C1-C4 alkyl, halo, oxo, CF3 or Phenyl; R5b is H, C1-C4 alkyl, aryl, heteroaryl, C3-C6 cycloalkyl, 4-7 membered heterocycloalkyl; and m5 is 0, 1, or 2.
32. The compound, or pharmaceutically acceptable salt thereof, according to claim 30; wherein Cy3 is selected from:
Figure imgf000269_0002
And wherein each R5a is independently C1-C4 alkyl, halo, oxo, CF3 or Ph; R5b is H, C1-C4 alkyl, aryl, heteroaryl, C3-C6 cycloalkyl, 4-7 membered heterocycloalkyl; R5c is H, CpC4 alkyl; m5 is 0, 1, or 2; and n5 is θ, I, or 2.
33. The compound according to claim 1 or 2 wherein the compound is selected from the compounds listed in Table 1.
34. The compound according to any one of claims 1 to 33 wherein the disease involves inflammation.
35. The compound according to any one of claims 1 to 33, wherein the disease is a condition involving an immune response or an autoimmune disease.
36. The compound according to any one of claims 1 to 33, wherein the disease involves an impairment of cartilage turnover.
37. The compound according to any one of claims 1 to 33, wherein the disease is a proliferative disease.
38. The compound according to any one of claims 1 to 33, wherein the condition is a congenital cartilage malformation.
39. The compound according to any one of claims 1 to 33, wherein the disease is associated with hypersecretion of IL6.
40. The compound according to any one of claims 1 to 33, wherein the condition is transplantation rejction.
41. A compound according to Formula X, or XI:
Figure imgf000270_0001
wherein R3b is substituted or unsubstituted 4-7 membered heterocycloalkyl; provided that when the compound is according to Formula X, the heterocycloalkyl ring is other than unsubstituted morpholin-1- yi.
42. The compound according to claim 41 wherein R3b is unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, azepinyl, pyrrolidonyl, pyranyl, dihydrothiophenyl, dihydropyranyl, dihydrofuranyl, dihydrothiazolyl, tetrahydrofuranyl, tetrahydrothiophenyl, dioxanyl, tetrahydropyranyl, imidazolinyl, imidazolidinonyl, oxazolinyl, thiazolinyl, 2-pyrazolineyl, pyrazolidinyl, thiomorpholinyl- S-oxide, and thiomorpholinyl-S,S-dioxide piperidonyl, or piperazonyl.
43. The compound according to claim 41 wherein R3b is unsubstituted azetidin-1-yl, pyrrolidin-1-yl, piperidin-1-yl, morpholin-1 -yl, thiomorpholin-l-yl-S,S-dioxide, piperazin-1-yl, or azepin-1-yl.
44. The compound according to claim 41 wherein R3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1- yl, morpholin-1 -yl, piperazin-1-yl, or azepin-1-yl; substituted with one or more group selected from Ci- C4 alkyl, C1-C4 haloalkyl, cyano, amino, dialkylamino, dialkylaminomethyl, hydroxy, halo, acyl, acylamino, C1-C4 hydroxyalkyl, C1-C4 alkoxy, carboxamido, and C1-C4 dialkyl carboxamido.
45. The compound according to claim 41 wherein R3b is azetidin-1-yl, pyrrolidin-1-yl, piperidin-1- yl, morpholin-1 -yl, piperazin-1-yl, or azepin-1-yl; substituted with Me, CF3, F, Cl, difluoro, dimethyl, hydroxy, cyano, dimethylamino, dimethylaminomethyl, hydroxymethyl, carboxamido, N,N- dimethylcarboxamido, methoxy, ethoxy, or 2,2,2,-trifluoroethyl.
PCT/EP2009/059604 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases WO2010010190A1 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
CA2730757A CA2730757A1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
JP2011519188A JP5559168B2 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
DK09781072.5T DK2361251T3 (en) 2008-07-25 2009-07-24 HIS UNKNOWN RELATIONSHIPS USED TO TREAT DEGENERATIVE AND INFLAMMATORY DISEASES
RS20140345A RS53406B (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
BRPI0916659A BRPI0916659B8 (en) 2008-07-25 2009-07-24 compounds useful for the treatment of degenerative and inflammatory diseases and their uses
SI200930973T SI2361251T1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
MEP-2014-72A ME02046B (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
US13/055,936 US8853240B2 (en) 2008-07-25 2009-07-24 Compounds useful for the treatment of degenerative and inflammatory diseases
EA201170257A EA018587B1 (en) 2008-07-25 2009-07-24 2-CYCLOPROPYLCARBONYLANINO-5-PHENYL[1,2,4]TRIAZOLO[1,5-a]PYRIDINE DERIVATIVES FOR THE TREATMENT OF DEGENERATIVE AND INFLAMMATORY DISEASES
CN200980129088.4A CN102105471B (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
AU2009273143A AU2009273143B2 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
KR1020117004383A KR101711911B1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
EP09781072.5A EP2361251B1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
MX2011000758A MX2011000758A (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases.
ES09781072.5T ES2485913T3 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
PL09781072T PL2361251T3 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
IL210261A IL210261A (en) 2008-07-25 2010-12-26 5-(substituted phenyl)-s-triazolo[1,5-a]pyridin-2-yl amides of cyclopropane carboxylic acid for use as medicaments
ZA2011/00396A ZA201100396B (en) 2008-07-25 2011-01-14 Novel compounds useful for the treatment of degenerative and inflammatory diseases
HK11112990.2A HK1158635A1 (en) 2008-07-25 2011-11-30 Novel compounds useful for the treatment of degenerative and inflammatory diseases
SM201400072T SMT201400072B (en) 2008-07-25 2014-06-17 New compounds useful for the treatment of degenerative and inflammatory diseases
HRP20140681AT HRP20140681T1 (en) 2008-07-25 2014-07-15 Novel compounds useful for the treatment of degenerative and inflammatory diseases
US14/507,396 US9415037B2 (en) 2008-07-25 2014-10-06 Compounds useful for the treatment of degenerative and inflammatory diseases
US15/236,578 US10206907B2 (en) 2008-07-25 2016-08-15 Compounds useful for the treatment of degenerative and inflammatory diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US13592008P 2008-07-25 2008-07-25
US61/135,920 2008-07-25
US22068509P 2009-06-26 2009-06-26
US61/220,685 2009-06-26

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/055,936 A-371-Of-International US8853240B2 (en) 2008-07-25 2009-07-24 Compounds useful for the treatment of degenerative and inflammatory diseases
US14/507,396 Division US9415037B2 (en) 2008-07-25 2014-10-06 Compounds useful for the treatment of degenerative and inflammatory diseases

Publications (1)

Publication Number Publication Date
WO2010010190A1 true WO2010010190A1 (en) 2010-01-28

Family

ID=41021059

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2009/059604 WO2010010190A1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases
PCT/EP2009/059605 WO2010010191A1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases.

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/059605 WO2010010191A1 (en) 2008-07-25 2009-07-24 Novel compounds useful for the treatment of degenerative and inflammatory diseases.

Country Status (36)

Country Link
US (5) US8853240B2 (en)
EP (2) EP2346864B1 (en)
JP (2) JP5480261B2 (en)
KR (2) KR101676391B1 (en)
CN (2) CN102105471B (en)
AR (1) AR072875A1 (en)
AU (2) AU2009273143B2 (en)
BR (2) BRPI0916659B8 (en)
CA (2) CA2730762C (en)
CL (1) CL2009001637A1 (en)
CO (1) CO6361925A2 (en)
CR (1) CR20110092A (en)
CY (2) CY1113945T1 (en)
DK (2) DK2361251T3 (en)
DO (1) DOP2011000022A (en)
EA (2) EA018080B1 (en)
ES (2) ES2406691T3 (en)
HK (2) HK1158635A1 (en)
HR (2) HRP20130357T1 (en)
IL (2) IL210262A (en)
JO (1) JO3041B1 (en)
MA (1) MA32551B1 (en)
ME (1) ME02046B (en)
MX (2) MX2011000756A (en)
MY (1) MY157615A (en)
NZ (1) NZ590646A (en)
PE (1) PE20100152A1 (en)
PL (2) PL2346864T3 (en)
PT (2) PT2346864E (en)
RS (2) RS52823B (en)
SI (2) SI2361251T1 (en)
SM (2) SMT201300051B (en)
TW (1) TWI447118B (en)
UY (1) UY32006A (en)
WO (2) WO2010010190A1 (en)
ZA (2) ZA201100396B (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010149769A1 (en) 2009-06-26 2010-12-29 Galapagos Nv 5-phenyl-[1,2,4 ]triazolo[1,5-a]pyridin-2-yl carboxamides as jak inhibitors
WO2012000970A1 (en) 2010-07-01 2012-01-05 Cellzome Limited Triazolopyridines as tyk2 inhibitors
CN102459261A (en) * 2009-06-26 2012-05-16 加拉帕戈斯股份有限公司 Novel compound useful for the treatment of degenerative and inflammatory diseases
WO2012146657A1 (en) * 2011-04-28 2012-11-01 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
EP2566333A1 (en) * 2010-05-07 2013-03-13 The Board of Trustees of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
US8501936B2 (en) 2009-06-05 2013-08-06 Cephalon, Inc. Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
WO2014118388A1 (en) 2013-02-04 2014-08-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for assaying jak2 activity in red blood cells and uses thereof
EP2934514A4 (en) * 2012-12-21 2016-06-08 Univ Leland Stanford Junior Transthyretin stabilizers and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
US9394301B2 (en) 2007-10-10 2016-07-19 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use
WO2017012770A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Acid addition salts of filgotinib
WO2017012773A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Solid forms of filgotinib free base
WO2017012771A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Crystalline filgotinib sulfonic acid addition salts
KR20170044202A (en) 2014-09-02 2017-04-24 니뽄 신야쿠 가부시키가이샤 Pyrazolothiazole compound and medicine comprising same
WO2017150477A1 (en) 2016-03-01 2017-09-08 日本新薬株式会社 Crystal of compound having jak-inhibiting activity
WO2018149925A1 (en) 2017-02-17 2018-08-23 Galapagos Nv Anti-inflammatory compositions comprising irak and jak inhibitors
WO2018167283A1 (en) 2017-03-17 2018-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma associated neural remodeling
WO2018189335A1 (en) 2017-04-13 2018-10-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma
US10172959B2 (en) 2014-08-14 2019-01-08 Mamoun M. Alhamadsheh Systems for stabilizing and delivering active agents
US10206907B2 (en) 2008-07-25 2019-02-19 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
EP3492469A4 (en) * 2016-07-26 2019-10-09 Tianjin Longbogene Pharmaceutical Co., Ltd. Compound as selective jak inhibitor, and salt and therapeutic use thereof
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions for the treatment of inflammatory disorders
US10513497B2 (en) 2017-02-17 2019-12-24 Eidos Therapeutics, Inc. Process for preparing AG-10, its intermediates, and salts thereof
WO2020092015A1 (en) 2018-11-02 2020-05-07 University Of Rochester Therapeutic mitigation of epithelial infection
US10708263B2 (en) 2014-02-07 2020-07-07 Galapagos Nv Salts and pharmaceutical compositions thereof for the treatment of inflammatory disorders
US11058668B2 (en) 2018-03-23 2021-07-13 Eidos Therapeutics, Inc. Methods of treating TTR amyloidosis using AG10
US11260047B2 (en) 2018-08-17 2022-03-01 Eidos Therapeutics, Inc. Formulations of AG10
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
WO2023138550A1 (en) * 2022-01-18 2023-07-27 盛世泰科生物医药技术(苏州)有限公司 Cyclopropanamide-containing compound and use thereof
US12005043B2 (en) 2022-01-18 2024-06-11 Eidos Therapeutics, Inc. Formulations of AG10

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100331222A1 (en) * 2009-06-26 2010-12-30 Basf Se Process for producing cellulose beads from solutions of cellulose in ionic liquid
KR102427777B1 (en) 2012-06-26 2022-08-01 델 마 파마슈티컬스 Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
CN103965114B (en) * 2013-01-28 2016-01-06 苏州泽璟生物制药有限公司 Deuterated phenyl amino pyrimidine compounds and comprise the pharmaceutical composition of this compound
WO2014202458A1 (en) * 2013-06-19 2014-12-24 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of inflammatory disorders
JP6820097B2 (en) 2013-12-20 2021-01-27 サギメット バイオサイエンシーズ インコーポレイテッド Heterocyclic modulators of lipid synthesis and their combinations
US20180002328A1 (en) 2015-01-28 2018-01-04 Jn Therapeutics Substituted imidazo[1, 2-a]pyridin-2-ylamine compounds, and pharmaceutical compositions and methods of use thereof
SG11201708181RA (en) * 2015-04-13 2017-11-29 Galapagos Nv Methods for the treatment of inflammatory disorders
PL3290418T3 (en) * 2015-04-29 2019-11-29 Wuxi Fortune Pharmaceutical Co Ltd Janus kinase (jak) inhibitors
AU2016257892A1 (en) * 2015-05-05 2017-11-09 Concert Pharmaceuticals, Inc. Deuterated filgotinib
CN105061420B (en) * 2015-06-04 2017-09-05 南京旗昌医药科技有限公司 A kind of crystal formation of JAK inhibitor and its preparation method and application
CN104987333B (en) * 2015-07-14 2017-01-11 苏州富士莱医药股份有限公司 Filgotinib synthetic method
CN105198876B (en) * 2015-09-18 2017-09-29 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative H crystal form and preparation method thereof
CN105198879B (en) * 2015-09-18 2017-09-22 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative C crystal form and preparation method thereof
CN105218539B (en) * 2015-09-18 2017-07-25 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative B crystal form and preparation method thereof
CN105198878B (en) * 2015-09-18 2017-07-25 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative F crystal formations and preparation method thereof
CN105198877B (en) * 2015-09-18 2017-09-22 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative G crystal formations and preparation method thereof
CN105198880B (en) * 2015-09-18 2017-07-25 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative A crystal formations and preparation method thereof
CN105111207B (en) * 2015-09-18 2017-07-25 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative form D and preparation method thereof
CN105111206B (en) * 2015-09-18 2017-07-25 上海宣创生物科技有限公司 A kind of cyclopropanecarbonyl amine derivative crystal form E and preparation method thereof
CN108349974B (en) * 2016-02-02 2021-04-30 深圳市塔吉瑞生物医药有限公司 Substituted pyridine amide compound and application thereof
JOP20190024A1 (en) 2016-08-26 2019-02-19 Gilead Sciences Inc Substituted pyrrolizine compounds and uses thereof
CN107880038B (en) * 2016-09-30 2021-09-28 中国医药研究开发中心有限公司 [1,2,4] triazolo [1,5-a ] pyridine compound and preparation method and medical application thereof
JOP20190077A1 (en) * 2016-10-10 2019-04-09 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
EA036903B1 (en) * 2016-10-26 2021-01-13 Янссен Фармацевтика Нв Fused bicylic pyridine compounds and their use as ampa receptor modulators
AU2017350688B2 (en) * 2016-10-26 2021-04-01 Rapport Therapeutics, Inc. Fused azaheterocyclic compounds and their use as AMPA receptor modulators
EA039352B1 (en) * 2017-01-19 2022-01-17 Сучжоу Лонгбайотек Фармасьютикалз Ко., Лтд. Compound as selective jak inhibitor and salts and therapeutic use thereof
CN108341814B (en) * 2017-01-23 2021-09-03 上海翔锦生物科技有限公司 JAK kinase inhibitors and uses thereof
US11427581B2 (en) 2018-01-31 2022-08-30 Zhuhai United Laboratories Co., Ltd. JAK inhibitor and use thereof
EP3759109B1 (en) 2018-02-26 2023-08-30 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
CA3090545A1 (en) * 2018-02-28 2019-09-06 University Of Southern California Compositions and methods for modulating inflammatory and degenerative disorder
RS65243B1 (en) * 2018-08-23 2024-03-29 Zhuhai United Laboratories Co Ltd [1,2,4]triazolo[1,5-a]pyridine compound as jak inhibitor and application thereof
US10815227B2 (en) 2018-08-27 2020-10-27 Cadila Healthcare Limited Processes for the preparation of filgotinib
WO2020097400A1 (en) * 2018-11-07 2020-05-14 Dana-Farber Cancer Institute, Inc. Imidazopyridine derivatives and aza-imidazopyridine derivatives as janus kinase 2 inhibitors and uses thereof
US11633399B2 (en) 2018-12-25 2023-04-25 Sol-Gel Technologies Ltd. Treatment of skin disorders with compositions comprising an EGFR inhibitor
CN111892592B (en) * 2019-08-06 2023-09-19 江苏柯菲平医药股份有限公司 JAK kinase inhibitors and uses thereof
CN111072655B (en) * 2019-12-30 2021-12-10 深圳市坤健创新药物研究院 Triazole pyridine compound, preparation method thereof, medicinal composition and application
CA3169832A1 (en) * 2020-02-13 2021-08-19 Zhuhai United Laboratories Co., Ltd. Use of jak inhibitors in preparation of drugs for treatment of jak kinase related diseases
CA3166743A1 (en) * 2020-02-21 2021-08-26 Weiwei Mao Crystalline forms of jak inhibitor and uses thereof
AU2021267373A1 (en) 2020-05-06 2022-12-08 Ajax Therapeutics, Inc. 6-heteroaryloxy benzimidazoles and azabenzimidazoles as JAK2 inhibitors
CN111617318B (en) * 2020-05-26 2021-12-10 南京市儿童医院 Preparation method of injectable epiphyseal plate regeneration hydrogel for children
CN114394965B (en) * 2021-01-29 2023-09-12 深圳市乐土生物医药有限公司 Triazolopyridine compound and preparation method and application thereof
AU2022388555A1 (en) 2021-11-09 2024-05-02 Ajax Therapeutics, Inc. 6-he tero aryloxy benzimidazoles and azabenzimidazoles as jak2 inhibitors
CN113773322B (en) * 2021-11-10 2022-02-11 奥锐特药业(天津)有限公司 Preparation method of Filgotinib
US11596612B1 (en) 2022-03-08 2023-03-07 PTC Innovations, LLC Topical anesthetics
US11746108B1 (en) * 2022-06-13 2023-09-05 Apm Therapeutics 1, Inc. Triazolopyridine polymorph A

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003010167A1 (en) * 2001-07-20 2003-02-06 F. Hoffmann-La Roche Ag 8-methoxy-(1,2,4)triazolo(1,5-a)pyridine derivatives and their use as adenosine receptor ligands
US20050222171A1 (en) * 2004-01-22 2005-10-06 Guido Bold Organic compounds
WO2007009773A1 (en) * 2005-07-21 2007-01-25 Novartis Ag Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors
WO2008025821A1 (en) * 2006-08-30 2008-03-06 Cellzome Limited Triazole derivatives as kinase inhibitors
WO2008150015A1 (en) * 2007-06-05 2008-12-11 Takeda Pharmaceutical Company Limited Heterobicyclic compounds as kinase inhibitors
WO2009010530A1 (en) * 2007-07-18 2009-01-22 Novartis Ag Bicyclic heteroaryl compounds and their use as kinase inhibitors
WO2009017954A1 (en) * 2007-08-01 2009-02-05 Phenomix Corporation Inhibitors of jak2 kinase
WO2009027283A1 (en) * 2007-08-31 2009-03-05 Merck Serono S.A. Triazolopyridine compounds and their use as ask inhibitors
WO2009047514A1 (en) * 2007-10-10 2009-04-16 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4058243B2 (en) 1999-01-29 2008-03-05 中外製薬株式会社 Cartilage formation promoter and indoline-2-one derivative
EP1391211A1 (en) 2001-04-27 2004-02-25 Chugai Seiyaku Kabushiki Kaisha Chondrogenesis promoters
WO2004072072A1 (en) 2003-02-14 2004-08-26 Pfizer Products Inc. Triazolo-pyridines as anti-inflammatory compounds
TWI221602B (en) * 2003-07-21 2004-10-01 Benq Corp Emergent disc-withdrawing apparatus
US20070004658A1 (en) 2004-06-21 2007-01-04 Nick Vandeghinste Method and means for treatment of osteoarthritis
MX2007002050A (en) 2004-08-18 2007-03-29 Pharmacia & Upjohn Co Llc Triazolopyridine compounds.
US7524860B2 (en) 2004-10-07 2009-04-28 Pfizer Inc. Antibacterial agents
ATE495743T1 (en) * 2006-12-01 2011-02-15 Galapagos Nv TRIAZOLOPYRIDINE COMPOUNDS FOR THE TREATMENT OF DEGENERATIONAL AND INFLAMMATORY DISEASES
US8811196B2 (en) * 2008-02-19 2014-08-19 Qualcomm Incorporated Providing remote field testing for mobile devices
US20090217316A1 (en) * 2008-02-22 2009-08-27 Binita Gupta Systems and Methods for Advertising Insertion Notification in a Real-Time Streaming Media Service
BRPI0909945A2 (en) 2008-06-20 2015-07-28 Genentech Inc "compound, pharmaceutical composition, method for treating or alleviating the severity of a disease or condition responsive to inhibition of jak2 kinase activity in a patient, kit for treating a disease or disorder responsive to inhibition of jak kinase"
WO2010010184A1 (en) 2008-07-25 2010-01-28 Galapagos Nv [1, 2, 4] triazolo [1, 5-a] pyridines as jak inhibitors
WO2010010186A1 (en) * 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010188A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010187A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
JO3041B1 (en) * 2008-07-25 2016-09-05 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010189A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
DE102009000543A1 (en) * 2009-02-02 2010-08-12 Evonik Degussa Gmbh Process, absorption media and apparatus for absorbing CO2 from gas mixtures
CN103961977A (en) * 2009-02-02 2014-08-06 巴斯夫欧洲公司 Cyclic-amine-comprisng absorption medium for removing acid gases
BRPI1009637A2 (en) 2009-06-05 2019-04-30 Cephalon, Inc compound, composition and use of a compound
JO3030B1 (en) 2009-06-26 2016-09-05 Galapagos Nv Novel Compound Useful for the Treatment of Degenerative and Inflammatory Diseases
TWI462920B (en) 2009-06-26 2014-12-01 葛萊伯格有限公司 Novel compound useful for the treatment of degenerative and inflammatory diseases
US20130310340A1 (en) 2012-05-16 2013-11-21 Rigel Pharmaceuticals, Inc. Method of treating muscular degradation
CN104379173A (en) 2012-06-22 2015-02-25 加拉帕戈斯股份有限公司 Aminotriazolopyridine for use in the treatment of inflammation, and pharmaceutical compositions thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003010167A1 (en) * 2001-07-20 2003-02-06 F. Hoffmann-La Roche Ag 8-methoxy-(1,2,4)triazolo(1,5-a)pyridine derivatives and their use as adenosine receptor ligands
US20050222171A1 (en) * 2004-01-22 2005-10-06 Guido Bold Organic compounds
WO2007009773A1 (en) * 2005-07-21 2007-01-25 Novartis Ag Pyrazolo[1.5-a]pyrimidin-7-yl amine derivatives as protein kinase inhibitors
WO2008025821A1 (en) * 2006-08-30 2008-03-06 Cellzome Limited Triazole derivatives as kinase inhibitors
WO2008150015A1 (en) * 2007-06-05 2008-12-11 Takeda Pharmaceutical Company Limited Heterobicyclic compounds as kinase inhibitors
WO2009010530A1 (en) * 2007-07-18 2009-01-22 Novartis Ag Bicyclic heteroaryl compounds and their use as kinase inhibitors
WO2009017954A1 (en) * 2007-08-01 2009-02-05 Phenomix Corporation Inhibitors of jak2 kinase
WO2009027283A1 (en) * 2007-08-31 2009-03-05 Merck Serono S.A. Triazolopyridine compounds and their use as ask inhibitors
WO2009047514A1 (en) * 2007-10-10 2009-04-16 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use

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 (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9394301B2 (en) 2007-10-10 2016-07-19 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use
US9771362B2 (en) 2007-10-10 2017-09-26 Cancer Research Technology Limited [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-c]pyrimidine compounds and their use
US10206907B2 (en) 2008-07-25 2019-02-19 Galapagos Nv Compounds useful for the treatment of degenerative and inflammatory diseases
US8633173B2 (en) 2009-06-05 2014-01-21 Cephalon, Inc Preparation and uses of 1,2,4-triazolo [1,5a] pyridine derivatives
US8501936B2 (en) 2009-06-05 2013-08-06 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
GB2483424A (en) * 2009-06-26 2012-03-07 Galapagos Nv 5-Phenyl-[1,2,4] triazolo[1,5-A] pyridin-2-yl carboxamides as JAK inhibitors
GB2483424B (en) * 2009-06-26 2012-07-18 Galapagos Nv 5-Phenyl-[1,2,4] triazolo[1,5-A]pyridin-2-yl carboxamides as jak inhibitors
US8088764B2 (en) 2009-06-26 2012-01-03 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
CN102459261A (en) * 2009-06-26 2012-05-16 加拉帕戈斯股份有限公司 Novel 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
KR20120107919A (en) * 2009-06-26 2012-10-04 갈라파고스 엔.브이. 5--12415--2- 5-phenyl-124triazolo15-apyridin-2-yl carboxamides as jak inhibitors
US10328081B2 (en) 2009-06-26 2019-06-25 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
KR101696669B1 (en) 2009-06-26 2017-01-16 갈라파고스 엔.브이. 5-phenyl-(1,2,4)triazolo(1,5-a)pyridin-2-yl carboxamides as jak inhibitors
EA021637B1 (en) * 2009-06-26 2015-07-30 Галапагос Нв 5-phenyl-[1,2,4]triazolo[1,5-a]pyridin-2-yl carboxamides as jak inhibitors
AU2010264633B2 (en) * 2009-06-26 2015-06-04 Galapagos Nv 5-phenyl-[1,2,4 ]triazolo[1,5-a]pyridin-2-yl carboxamides as JAK inhibitors
EP3178819A1 (en) 2009-06-26 2017-06-14 Galapagos NV 5-phenyl-[1,2,4]triazolo[1,5-a]pyridin-2-yl carboxamides as jak inhibitors
US8796457B2 (en) 2009-06-26 2014-08-05 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
WO2010149769A1 (en) 2009-06-26 2010-12-29 Galapagos Nv 5-phenyl-[1,2,4 ]triazolo[1,5-a]pyridin-2-yl carboxamides as jak inhibitors
EP2445911B1 (en) 2009-06-26 2017-03-01 Galapagos N.V. 5-phenyl-[1,2,4]triazolo[1,5-a]pyridin-2-yl carboxamides as jak inhibitors
EP2445912B1 (en) * 2009-06-26 2014-08-13 Galapagos N.V. Novel compound useful for the treatment of degenerative and inflammatory diseases
AU2010264635B2 (en) * 2009-06-26 2015-05-07 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
CN102459261B (en) * 2009-06-26 2014-11-05 加拉帕戈斯股份有限公司 Novel compound useful for the treatment of degenerative and inflammatory diseases
US10278929B2 (en) 2010-05-07 2019-05-07 The Board Of Trustees Of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
US11337935B2 (en) 2010-05-07 2022-05-24 The Board Of Trustees Of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
US9308209B2 (en) 2010-05-07 2016-04-12 The Board Of Trustees Of The Leland Stanford Junio Identification of stabilizers of multimeric proteins
US8877795B2 (en) 2010-05-07 2014-11-04 The Board Of Trustees Of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
EP2566333A4 (en) * 2010-05-07 2014-04-02 Univ Leland Stanford Junior Identification of stabilizers of multimeric proteins
EP2566333A1 (en) * 2010-05-07 2013-03-13 The Board of Trustees of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
US10039726B2 (en) 2010-05-07 2018-08-07 The Board Of Trustees Of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
WO2012000970A1 (en) 2010-07-01 2012-01-05 Cellzome Limited Triazolopyridines as tyk2 inhibitors
CN103492386A (en) * 2011-04-28 2014-01-01 加拉帕戈斯股份有限公司 Novel compound useful for the treatment of degenerative and inflammatory diseases
CN103492385A (en) * 2011-04-28 2014-01-01 加拉帕戈斯股份有限公司 Novel compound useful for the treatment of degenerative and inflammatory diseases
WO2012146657A1 (en) * 2011-04-28 2012-11-01 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
CN103492386B (en) * 2011-04-28 2016-03-09 加拉帕戈斯股份有限公司 Can be used for the new compound for the treatment of sex change and inflammatory diseases
EA024743B1 (en) * 2011-04-28 2016-10-31 Галапагос Нв Compound useful for the treatment of degenerative and inflammatory conditions
US9241931B2 (en) 2011-04-28 2016-01-26 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
US8975406B2 (en) 2011-04-28 2015-03-10 Galapagos N.V. Compound useful for the treatment of degenerative and inflammatory diseases
AU2012247482B2 (en) * 2011-04-28 2015-03-05 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
AU2012247484B2 (en) * 2011-04-28 2015-02-19 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
WO2012146659A1 (en) * 2011-04-28 2012-11-01 Galapagos Nv Novel compound useful for the treatment of degenerative and inflammatory diseases
US8802682B2 (en) 2011-04-28 2014-08-12 Galapagos Nv Compound useful for the treatment of degenerative and inflammatory diseases
KR101934707B1 (en) 2011-04-28 2019-01-03 갈라파고스 엔.브이. Novel compound useful for the treatment of degenerative and inflammatory diseases
JP2014512405A (en) * 2011-04-28 2014-05-22 ガラパゴス・ナムローゼ・フェンノートシャップ Novel compounds useful for the treatment of degenerative and inflammatory diseases
US9987272B2 (en) 2011-04-28 2018-06-05 Galapagos Nv Compound useful for the treatement of degenerative and inflammatory diseases
JP2014512404A (en) * 2011-04-28 2014-05-22 ガラパゴス・ナムローゼ・フェンノートシャップ Novel compounds useful for the treatment of degenerative and inflammatory diseases
CN103492385B (en) * 2011-04-28 2016-05-11 加拉帕戈斯股份有限公司 Be used for the treatment of the noval chemical compound of degenerative disease and inflammatory disease
US9913826B2 (en) 2012-12-21 2018-03-13 The Board Of Trustees Of The Leland Stanford Junior University Compounds and compositions that bind and stabilize transthyretin and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
US10398681B2 (en) 2012-12-21 2019-09-03 The Board Of Trustees Of The Leland Stanford Junior University Compounds and compositions that bind and stabilize transthyretin and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
US10842777B2 (en) 2012-12-21 2020-11-24 The Board Of Trustees Of The Leland Stanford Junior University Compounds and compositions that bind and stabilize transthyretin and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
EP2934514A4 (en) * 2012-12-21 2016-06-08 Univ Leland Stanford Junior Transthyretin stabilizers and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
WO2014118388A1 (en) 2013-02-04 2014-08-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for assaying jak2 activity in red blood cells and uses thereof
US10493158B2 (en) 2014-02-07 2019-12-03 Galapagos Nv Pharmaceutical compositions 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
US11667633B2 (en) 2014-02-07 2023-06-06 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
US10172959B2 (en) 2014-08-14 2019-01-08 Mamoun M. Alhamadsheh Systems for stabilizing and delivering active agents
US11129902B2 (en) 2014-08-14 2021-09-28 Mamoun M. Alhamadsheh Enhanced SN-38 anticancer agent
US10363318B2 (en) 2014-08-14 2019-07-30 Mamoun M. Alhamadsheh Enhanced active agents
US10596269B2 (en) 2014-08-14 2020-03-24 Mamoun M. Alhamadsheh Delivering enhanced active agents
US10772967B2 (en) 2014-08-14 2020-09-15 Mamoun M. Alhamadsheh Enhanced anticancer agent
US9999622B2 (en) 2014-09-02 2018-06-19 Nippon Shinyaku Co., Ltd. Pyrazolothiazole compound and medicine comprising same
US9937176B2 (en) 2014-09-02 2018-04-10 Nippon Shinyaku Co., Ltd. Pyrazolothiazole compound and medicine comprising same
KR20170044202A (en) 2014-09-02 2017-04-24 니뽄 신야쿠 가부시키가이샤 Pyrazolothiazole compound and medicine comprising same
WO2017012773A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Solid forms of filgotinib free base
WO2017012770A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Acid addition salts of filgotinib
WO2017012771A1 (en) 2015-07-23 2017-01-26 Ratiopharm Gmbh Crystalline filgotinib sulfonic acid addition salts
US10822350B2 (en) 2016-03-01 2020-11-03 Nippon Shinyaku Co., Ltd. Crystal of compound having JAK-inhibiting activity
WO2017150477A1 (en) 2016-03-01 2017-09-08 日本新薬株式会社 Crystal of compound having jak-inhibiting activity
KR20180116341A (en) 2016-03-01 2018-10-24 니뽄 신야쿠 가부시키가이샤 Determination of compounds having JAK inhibiting action
US11377453B2 (en) 2016-03-01 2022-07-05 Nippon Shinyaku Co., Ltd. Crystal of compound having JAK-inhibiting activity
US11414413B2 (en) 2016-07-26 2022-08-16 Suzhou Longbiotech Pharmaceuticals Co., Ltd. Heterocyclic compound as JAK inhibitor, and salts and therapeutic use thereof
EP3492469A4 (en) * 2016-07-26 2019-10-09 Tianjin Longbogene Pharmaceutical Co., Ltd. Compound as selective jak inhibitor, and salt and therapeutic use thereof
US11279699B2 (en) 2016-07-26 2022-03-22 Suzhou Longbiotech Pharmaceuticals Co., Ltd. Compound as selective JAK inhibitor, and salt and therapeutic use thereof
US11919865B2 (en) 2017-02-17 2024-03-05 Eidos Therapeutics, Inc. Processes for preparing AG-10, its intermediates, and salts thereof
US11078162B2 (en) 2017-02-17 2021-08-03 Eidos Therapeutics, Inc. Processes for preparing AG-10, its intermediates, and salts thereof
WO2018149925A1 (en) 2017-02-17 2018-08-23 Galapagos Nv Anti-inflammatory compositions comprising irak and jak inhibitors
US10513497B2 (en) 2017-02-17 2019-12-24 Eidos Therapeutics, Inc. Process for preparing AG-10, its intermediates, and salts thereof
WO2018167283A1 (en) 2017-03-17 2018-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma associated neural remodeling
WO2018189335A1 (en) 2017-04-13 2018-10-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the diagnosis and treatment of pancreatic ductal adenocarcinoma
US11058668B2 (en) 2018-03-23 2021-07-13 Eidos Therapeutics, Inc. Methods of treating TTR amyloidosis using AG10
US11260047B2 (en) 2018-08-17 2022-03-01 Eidos Therapeutics, Inc. Formulations of AG10
WO2020092015A1 (en) 2018-11-02 2020-05-07 University Of Rochester Therapeutic mitigation of epithelial infection
US11427558B1 (en) 2019-07-11 2022-08-30 ESCAPE Bio, Inc. Indazoles and azaindazoles as LRRK2 inhibitors
WO2023138550A1 (en) * 2022-01-18 2023-07-27 盛世泰科生物医药技术(苏州)有限公司 Cyclopropanamide-containing compound and use thereof
US12005043B2 (en) 2022-01-18 2024-06-11 Eidos Therapeutics, Inc. Formulations of AG10

Also Published As

Publication number Publication date
US20110190260A1 (en) 2011-08-04
AU2009273144B2 (en) 2013-10-31
PL2346864T3 (en) 2013-08-30
US20100029709A1 (en) 2010-02-04
CA2730757A1 (en) 2010-01-28
IL210261A (en) 2017-03-30
BRPI0916659A2 (en) 2019-03-12
CA2730762A1 (en) 2010-01-28
EP2346864A1 (en) 2011-07-27
IL210262A (en) 2014-09-30
US8242274B2 (en) 2012-08-14
US20170035738A1 (en) 2017-02-09
MY157615A (en) 2016-06-30
EA201170255A1 (en) 2011-08-30
HRP20130357T1 (en) 2013-05-31
CN102105472B (en) 2013-04-17
IL210262A0 (en) 2011-03-31
JO3041B1 (en) 2016-09-05
CY1115569T1 (en) 2017-01-04
CR20110092A (en) 2011-03-16
BRPI0916862B8 (en) 2021-05-25
CO6361925A2 (en) 2012-01-20
BRPI0916659B1 (en) 2020-12-08
SMT201300051B (en) 2013-07-09
DK2361251T3 (en) 2014-07-21
JP2011529033A (en) 2011-12-01
US9415037B2 (en) 2016-08-16
US8853240B2 (en) 2014-10-07
RS53406B (en) 2014-10-31
JP2011529032A (en) 2011-12-01
DOP2011000022A (en) 2011-03-31
CN102105472A (en) 2011-06-22
BRPI0916862B1 (en) 2020-08-11
CY1113945T1 (en) 2016-07-27
PT2346864E (en) 2013-04-04
JP5480261B2 (en) 2014-04-23
TW201008939A (en) 2010-03-01
SI2346864T1 (en) 2013-05-31
CL2009001637A1 (en) 2010-02-19
PE20100152A1 (en) 2010-03-24
UY32006A (en) 2010-02-26
WO2010010191A1 (en) 2010-01-28
HK1158634A1 (en) 2012-07-20
ZA201100396B (en) 2011-10-26
PL2361251T3 (en) 2014-10-31
KR101711911B1 (en) 2017-03-03
ME02046B (en) 2015-05-20
JP5559168B2 (en) 2014-07-23
ZA201100472B (en) 2011-10-26
PT2361251E (en) 2014-07-11
NZ590646A (en) 2011-11-25
ES2485913T3 (en) 2014-08-14
RS52823B (en) 2013-10-31
US20150150856A1 (en) 2015-06-04
US20120309784A1 (en) 2012-12-06
AU2009273143B2 (en) 2014-01-23
EP2346864B1 (en) 2013-03-06
ES2406691T3 (en) 2013-06-07
KR20110045020A (en) 2011-05-03
HRP20140681T1 (en) 2014-10-24
CN102105471A (en) 2011-06-22
KR101676391B1 (en) 2016-11-15
EA018587B1 (en) 2013-09-30
AU2009273143A1 (en) 2010-01-28
AU2009273144A1 (en) 2010-01-28
US10206907B2 (en) 2019-02-19
BRPI0916659B8 (en) 2021-05-25
MX2011000758A (en) 2011-02-23
SMT201400072B (en) 2014-09-08
MX2011000756A (en) 2011-03-21
EA201170257A1 (en) 2011-08-30
SI2361251T1 (en) 2014-09-30
TWI447118B (en) 2014-08-01
EA018080B1 (en) 2013-05-30
EP2361251B1 (en) 2014-04-30
AR072875A1 (en) 2010-09-29
DK2346864T3 (en) 2013-05-13
CA2730762C (en) 2017-11-21
HK1158635A1 (en) 2012-07-20
IL210261A0 (en) 2011-03-31
CN102105471B (en) 2014-10-15
EP2361251A1 (en) 2011-08-31
MA32551B1 (en) 2011-08-01
BRPI0916862A2 (en) 2016-02-10
KR20110053435A (en) 2011-05-23

Similar Documents

Publication Publication Date Title
AU2009273144B2 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010184A1 (en) [1, 2, 4] triazolo [1, 5-a] pyridines as jak inhibitors
CA2833963C (en) Novel compound useful for the treatment of degenerative and inflammatory diseases
WO2010010186A1 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010189A1 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010188A1 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010187A1 (en) Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2013117645A1 (en) Imidazo [4, 5 -c] pyridine derivatives useful for the treatment of degenerative and inflammatory diseases

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980129088.4

Country of ref document: CN

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

Ref document number: 09781072

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 210261

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2730757

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009273143

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/000758

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2011519188

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13055936

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2009273143

Country of ref document: AU

Date of ref document: 20090724

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 988/DELNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2009781072

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20117004383

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201170257

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: P-2014/0345

Country of ref document: RS

ENP Entry into the national phase

Ref document number: PI0916659

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110125