US20180298015A1 - PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS - Google Patents

PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS Download PDF

Info

Publication number
US20180298015A1
US20180298015A1 US16/014,231 US201816014231A US2018298015A1 US 20180298015 A1 US20180298015 A1 US 20180298015A1 US 201816014231 A US201816014231 A US 201816014231A US 2018298015 A1 US2018298015 A1 US 2018298015A1
Authority
US
United States
Prior art keywords
alkyl
halogen
optionally substituted
oxo
cycloalkyl group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/014,231
Other languages
English (en)
Inventor
Marian C. Bryan
Alberto Gobbi
James Richard Kiefer, JR.
Aleksandr Kolesnikov
Alan G. Olivero
Joy Drobnick
Jun Liang
Naomi Rajapaksa
Chudi Ndubaku
Jianwen Feng
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US16/014,231 priority Critical patent/US20180298015A1/en
Publication of US20180298015A1 publication Critical patent/US20180298015A1/en
Priority to US16/775,650 priority patent/US10988478B1/en
Priority to US17/172,766 priority patent/US20220332718A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • This invention pertains to compounds useful for inhibition of Interleukin-1 Receptor Associated Kinase 4 (IRAK4).
  • IRAK4 Interleukin-1 Receptor Associated Kinase 4
  • TIR-domain Toll-Interleukin 1 Receptor-domain
  • TLR Toll-like receptors
  • IL-1 and IL-18 receptors play critical roles in innate immunity and have been implicated in the pathogenesis of autoimmunity.
  • TLRs for example, recognize pathogenic or endogenous ligands and provide a requisite signal for dendritic cell maturation and antigen presentation to T cell.
  • proteins that mediate signaling from these receptors have also been shown to play important roles in the pathogenesis of autoimmune disorders.
  • mice deficient in MyD88 an adaptor protein that directly interacts with the TIR domain, are more susceptible to bacterial, fungal and parasitic infections.
  • MyD88 deficient mice are resistant to experimental autoimmune encephalomyelitis (EAE) and streptococcal cell wall-induced arthritis.
  • EAE experimental autoimmune encephalomyelitis
  • the Interleukin-1 Receptor Associated Kinase (IRAK) family is comprised of four family members IRAK1, IRAK2, IRAK3 (also termed IRAK-M), and IRAK4. These proteins are characterized by a typical N-terminal death domain that mediates interaction with MyD88-family adaptor proteins and a centrally located kinase domain. Whereas IRAK1 and IRAK4 have kinase activity, IRAK2 and IRAK3 are catalytically inactive. Upon activation of their upstream cognate receptors, IRAK4 is thought to phosphorylate IRAK1, resulting in the activation and autophosphorylation of IRAK1 and subsequent phosphorylation of downstream substrates.
  • IRAK1 The hyperphosphorylation of IRAK1 directs its dissociation from the receptor complex and its eventual ubiquitylation and proteasomal degradation. Phosphorylation of downstream substrates such as Pellino-2 ultimately leads to the activation of the MAPKs such as p38 and c-Jun N-terminal kinase (JNK) and NF-kB followed by production of pro-inflammatory cytokines, chemokines, and destructive enzyme.
  • MAPKs such as p38 and c-Jun N-terminal kinase (JNK) and NF-kB followed by production of pro-inflammatory cytokines, chemokines, and destructive enzyme.
  • IRAK4 A novel member of the IRAK family with the properties of an IRAK-kinase
  • PNAS 2002 99(8), 5567-5572
  • Flannery et al. “The interleukin-1 receptor-associated kinases: Critical regulators of innate immune signaling,” Biochem Pharm 2010, 80(12), 1981-1991.
  • Patients with destabilizing or null mutations in IRAK4 demonstrate defects in TLR signaling and the production of pro-inflammatory cytokines such as IL-1 and TNF as well as antiviral cytokines such as IFN ⁇ and IFN ⁇ .
  • IRAK4 deficient mice have defects in TLR- and IL-1-mediated cytokine production and exhibit an increased susceptibility to infection.
  • IRAK1 deficient mice demonstrate a loss of responsiveness to lipopolysaccharides (LPS), IL-1, and IL-18 as well as impaired Th1 development. These mice were resistant to experimental autoimmune encephalomyelitis, exhibiting little or no CNS inflammation.
  • compounds that modulate the function of IRAK4 represent an attractive approach to the development of therapeutic agents for the treatment of diseases such as inflammatory, cell proliferative and immune-related conditions and diseases associated with IRAK-mediated signal transduction, such as rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, lupus, diabetes, obesity, allergic disease, psoriasis, asthma, graft rejection, cancer and sepsis.
  • diseases such as inflammatory, cell proliferative and immune-related conditions and diseases associated with IRAK-mediated signal transduction, such as rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, lupus, diabetes, obesity, allergic disease, psoriasis, asthma, graft rejection, cancer and sepsis.
  • One aspect of the invention includes a compound of Formula 0:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 12 NR 6 R 7 , —NR 8 R 9 , —O—C 1-3 alkyl, a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group, heterocyclic group, heteroaryl ring, or aryl ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 12 NR 10 R 11 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, —C(O)NR 8 R 9 , or —NR 8 R 9 ,
  • any alkyl, alkenyl, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-7 membered monocyclic saturated or partially saturated heterocyclic group that may be optionally substituted with oxo;
  • R 5 is hydrogen, —CN, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —C(O)NR 8 R 9 , —O(C 3-7 cycloalkyl group), —O(C 1-3 alkyl)-3-8 membered cycloalkyl group, —O(C 0 -C 3 alkyl)-3-8 membered saturated or partially saturated heterocyclic group, —O(C 1-3 alkyl)-phenyl, a —O(C 1-3 alkyl)-5-6 membered heteroaryl ring, a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-11 membered saturated or partially saturated heterocyclic group that may be optionally substituted with (i) —C(O)(C 1-3 alkyl) optionally substituted with halogen or (ii) with C 1-3 alkyl optionally substituted with halogen, and
  • any cycloalkyl group, heterocyclic group, phenyl, or heteroaryl ring is optionally substituted by halogen; oxo; CN; OH; C 1-6 alkoxy; —NR 8 R 9 ; —C(O)(C 1-3 alkyl); —(C 0-3 alkyl)C(O)NR 10 R 11 ; —S(O) 12 NR 8 R 9 ; —OP(O)(OC 1-3 alkyl) 2 ; C 3-10 cycloalkyl group optionally substituted with OH or halogen; a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted with oxo or C 1-3 alkyl; a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl; or C 1-4 alkyl optionally substituted by halogen, oxo
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, —(C 0-3 alkyl)-C 3-6 cycloalkyl group, a —(C 0-3 alkyl)-3-11 membered heterocyclic group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 1-2 R 13 , —C(O)(C 1-3 alkyl), —C(O)NR 10 R 11 , —C(O)OR 13 , —S(O) 1-2 N 10 R 11 , or —(C 0-3 alkyl)-OP(O)(OC 1-3 alkyl) 2 ,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen; oxo; CN; OR 13 ; C 1-3 haloalkoxy; —C(O)(C 1-3 alkyl); —S—C 1-3 alkyl; or C 1-3 alkyl optionally substituted with OH, halogen, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-8 membered heterocyclic group, and
  • A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, —(C 0-3 alkyl)-phenyl, a 3-11 membered saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 1-2 R 13 , or R 10 and R 11 are taken together to form a 5-8 membered heterocyclic group,
  • any alkyl, cycloalkyl group, phenyl, heterocyclic group, or heteroaryl ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , —NR 6 R 7 , or a 5-6 membered monocyclic heteroaryl ring;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is hydrogen, halogen, CN, or C 1-3 alkyl optionally substituted with —NH 2 , halogen, or CN.
  • Another aspect of the invention includes a compound of Formula I:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 12 NR 6 R 7 , —NR 8 R 9 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and any cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 12 NR 10 R 11 , or —NR 8 R 9 ;
  • any alkyl or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —O(C 3-7 cycloalkyl group), —O(C 1-3 alkyl)-3-8 membered cycloalkyl group, —O(C 1-3 alkyl)-3-8 membered saturated or partially saturated heterocyclic group, —O(C 1-3 alkyl)-phenyl, a —O(C 1-3 alkyl)-5-6 membered heteroaryl ring, a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is optionally substituted by halogen, oxo, CN, OH, C 1-6 alkoxy, —C(O)(C 1-3 alkyl), —(C 0 -C 3 alkyl)C(O)NR 10 R 11 , —S(O) 12 NR 8 R 9 , —OP(O)(OC 1-3 alkyl) 2 , a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl, or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, phenyl, a 3-8 membered saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or —NR 8 R 9 ;
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, —(C 0 -C 3 alkyl)-C 3-6 cycloalkyl group, a —(C 0 -C 3 alkyl)-3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)(C 1-3 alkyl), —C(O)NR 10 R 11 , —C(O)OR 13 , —S(O) 1-2 NR 10 R 11 , or —OP(O)(OC 1-3 alkyl) 2 ,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-8 membered heterocyclic group;
  • A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, —(C 0-3 alkyl)-phenyl, a 3-11 membered saturated heterocyclic group, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 1-2 R 13 , or R 10 and R 11 are taken together to form a 5-8 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , or —NR 6 R 7 ;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is H, —Cl, —CN, or —CH 3 .
  • Yet another aspect of the invention includes a compound of Formula II:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 12 NR 6 R 7 , —NR 8 R 9 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and any cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 12 NR 10 R 11 , or —NR 8 R 9 ;
  • any alkyl or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —O(C 3-7 cycloalkyl group), a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is optionally substituted by halogen, oxo, CN, OH, —(C 0-3 alkyl)C(O)NR 10 R 11 , —OP(O)(OC 1-3 alkyl) 2 , or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, or —NR 8 R 9 ;
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , or —S(O) 1-2 NR 10 R 11 ,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy; wherein when A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, a 3-11 membered saturated heterocyclic group, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 1-2 R 13
  • any alkyl, cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , or —NR 6 R 7 ;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is H, —Cl, —CN, or —CH 3 .
  • composition that comprises a compound of the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • Another aspect includes a compound of the invention for use in therapy, such as the treatment of an inflammatory disease, an autoimmune disease or cancer.
  • Another aspect includes a method of preventing, treating or lessening the severity of a disease or condition responsive to the inhibition of IRAK4, in a patient.
  • the method can comprise administering to the patient a therapeutically effective amount of a compound of the invention.
  • Another aspect includes the use of a compound of the invention in the manufacture of a medicament for the treatment of a disease responsive to the inhibition of IRAK4.
  • kits for treating a disease or disorder responsive to the inhibition of IRAK4 can comprise a first pharmaceutical composition comprising a compound of the invention, and instructions for use.
  • Halogen or “halo” refers to F, Cl, Br or I. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical, wherein the alkyl radical may be optionally substituted.
  • the alkyl radical is one to eighteen carbon atoms (C 1 -C 18 ).
  • the alkyl radical is C 0 -C 6 , C 0 -C 5 , C 0 -C 3 , C 1 -C 12 , C 1 -C 10 , C 1 -C 8 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , or C 1 -C 3 .
  • C 0 alkyl refers to a bond.
  • alkyl groups include methyl (Me, —CH 3 ), ethyl (Et, —CH 2 CH 3 ), 1-propyl (n-Pr, n-propyl, —CH 2 CH 2 CH 3 ), 2-propyl (i-Pr, i-propyl, —CH(CH 3 ) 2 ), 1-butyl (n-Bu, n-butyl, —CH 2 CH 2 CH 2 CH 3 ), 2-methyl-1-propyl (i-Bu, i-butyl, —CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, —CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, —C(CH 3 ) 3 ), 1-pentyl (n-pentyl, —CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (—CH(CH 3 )CH 2 CH 2 CH 2 CH
  • substituents for “optionally substituted alkyls” include one to four instances of F, Cl, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(O)CH 3 , COOH, CO 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, SO 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.
  • Aryl refers to a carbocyclic aromatic group, whether or not fused to one or more groups, having the number of carbon atoms designated, or if no number is designated, up to 14 carbon atoms.
  • One example includes aryl groups having 6-14 carbon atoms.
  • Another example includes aryl groups having 6-10 carbon atoms.
  • Examples of aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, 1,2,3,4-tetrahydronaphthalenyl, 1H-indenyl, 2,3-dihydro-1H-indenyl, and the like (see, e.g., Lang's Handbook of Chemistry (Dean, J.
  • a particular aryl is phenyl.
  • Substituted phenyl or substituted aryl means a phenyl group or aryl group substituted with one, two, three, four or five substituents, for example, 1-2, 1-3 or 1-4 substituents, such as chosen from groups specified herein (see “optionally substituted” definition), such as F, Cl, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(O)CH 3 , COOH, CO 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesul
  • substituted phenyl examples include a mono- or di(halo)phenyl group such as 2-chlorophenyl, 2-bromophenyl, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5-dichlorophenyl, 3,4-dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3,4-dibromophenyl, 3-chloro-4-fluorophenyl, 2-fluorophenyl, 2,4-difluorophenyl and the like; a mono- or di(hydroxy)phenyl group such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4-dihydroxyphenyl, the protected-hydroxy derivatives thereof and the like; a nitrophenyl group such as 3- or 4-nitrophenyl; a cyanophenyl group, for example, 4-cyanophenyl; a mono- or di(al)phenyl group such as
  • substituted phenyl represents disubstituted phenyl groups where the substituents are different, for example, 3-methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4-ethyl-2-hydroxyphenyl, 3-hydroxy-4-nitrophenyl, 2-hydroxy-4-chlorophenyl, 2-chloro-5-difluoromethoxy and the like, as well as trisubstituted phenyl groups where the substituents are different, for example 3-methoxy-4-benzyloxy-6-methyl sulfonylamino, 3-methoxy-4-benzyloxy-6-phenyl sulfonylamino, and tetrasubstituted phenyl groups where the substituents are different such as 3-methoxy-4-benzyloxy-5-methyl-6-phenyl sulfonylamino.
  • compound(s) of the invention include compounds of Formula 0, Formula I, Formula II, and the compounds of Tables 1, 2 and 3 herein, including stereoisomers (including atropisomers), geometric isomers, tautomers, solvates, metabolites, isotopes, salts (e.g., pharmaceutically acceptable salts), and prodrugs thereof.
  • stereoisomers including atropisomers
  • geometric isomers including atropisomers
  • tautomers solvates, metabolites, isotopes, salts (e.g., pharmaceutically acceptable salts), and prodrugs thereof.
  • solvates, metabolites, isotopes or prodrugs are excluded, or any combination thereof.
  • Cycloalkyl refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein.
  • the cycloalkyl group is 3 to 12 carbon atoms (C 3 -C 12 ).
  • cycloalkyl is C 3 -C 8 , C 3 -C 10 or C 5 -C 10 .
  • the cycloalkyl group, as a monocycle is C 3 -C 8 , C 3 -C 6 or C 5 -C 6 .
  • the cycloalkyl group, as a bicycle is C 7 -C 12 .
  • the cycloalkyl group is C 5 -C 12 .
  • monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, perdeuteriocyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl.
  • Exemplary arrangements of bicyclic cycloalkyls having 7 to 12 ring atoms include, but are not limited to, [4,4], [4,5], [5,5], [5,6] or [6,6] ring systems.
  • Exemplary bridged bicyclic cycloalkyls include, but are not limited to, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane.
  • Examples of spiro cycloalkyl include, spiro[2.2]pentane, spiro[2.3]hexane, spiro[2.4]heptane, spiro[2.5]octane and spiro[4.5]decane.
  • substituents for “optionally substituted cycloalkyls” include one to four instances of F, Cl, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(O)CH 3 , COOH, CO 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, SO 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, aryl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.
  • Heterocyclic group “heterocyclic”, “heterocycle”, “heterocyclyl”, or “heterocyclo” are used interchangeably and refer to any mono-, bi-, tricyclic or spiro, saturated, partially saturated or unsaturated, aromatic (heteroaryl) or non-aromatic (e.g., heterocycloalkyl), ring system, having 3 to 20 ring atoms, where the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 3-11 ring atoms (“members”) and includes monocycles, bicycles, tricycles and spiro ring systems, wherein the ring atoms are carbon, where at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 1 to 4 heteroatoms. In one example, heterocyclyl includes 1 to 3 heteroatoms.
  • heterocyclyl includes 3- to 7-membered monocycles having 1-2, 1-3 or 1-4 heteroatoms selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 4- to 6-membered monocycles having 1-2, 1-3 or 1-4 heteroatoms selected from nitrogen, sulfur or oxygen.
  • heterocyclyl includes 3-membered monocycles.
  • heterocyclyl includes 4-membered monocycles.
  • heterocyclyl includes 5-6 membered monocycles, e.g., 5-6 membered heteroaryl.
  • heterocyclyl includes 3-11 membered heterocycloyalkyls, such as 4-11 membered heterocycloalkyls.
  • a heterocycloalkyl includes at least one nitrogen.
  • the heterocyclyl group includes 0 to 3 double bonds. Any nitrogen or sulfur heteroatom may optionally be oxidized (e.g., NO, SO, SO 2 ), and any nitrogen heteroatom may optionally be quaternized (e.g., [NR 4 ] + Cl ⁇ , [NR 4 ] + OH ⁇ ).
  • Example heterocycles are oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1,2-dithietanyl, 1,3-dithietanyl, pyrrolidinyl, dihydro-1H-pyrrolyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl, isoquinolinyl, tetrahydroisoquinolinyl, morpholinyl, thiomorpholinyl, 1,1-dioxo-thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, thi
  • Examples of 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms are thiazolyl, including thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, including 1,3,4-thiadiazol-5-yl and 1,2,4-thiadiazol-5-yl, oxazolyl, for example oxazol-2-yl, and oxadiazolyl, such as 1,3,4-oxadiazol-5-yl, and 1,2,4-oxadiazol-5-yl.
  • Example 5-membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as 1,3,4-triazol-5-yl; 1,2,3-triazol-5-yl, 1,2,4-triazol-5-yl, and tetrazolyl, such as 1H-tetrazol-5-yl.
  • Example benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl and benzimidazol-2-yl.
  • Example 6-membered heterocycles contain one to three nitrogen atoms and optionally a sulfur or oxygen atom, for example pyridyl, such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl; pyrimidyl, such as pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as 1,3,4-triazin-2-yl and 1,3,5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl, and pyrazinyl.
  • pyridyl such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl
  • pyrimidyl such as pyrimid-2-yl and pyrimid-4-yl
  • triazinyl such as 1,3,4-triazin-2-yl and 1,3,5-triazin-4-yl
  • pyridine N-oxides and pyridazine N-oxides and the pyridyl, pyrimid-2-yl, pyrimid-4-yl, pyridazinyl and the 1,3,4-triazin-2-yl groups are other example heterocycle groups. Heterocycles may be optionally substituted.
  • substituents for “optionally substituted heterocycles” include one to four instances of F, Cl, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(O)CH 3 , COOH, CO 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, SO 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, aryl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.
  • Heteroaryl refers to any mono-, bi-, or tricyclic ring system where at least one ring is a 5- or 6-membered aromatic ring containing from 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur, and in an example embodiment, at least one heteroatom is nitrogen. See, for example, Lang's Handbook of Chemistry (Dean, J. A., ed.) 13 th ed. Table 7-2 [1985]. Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to an aryl ring, wherein the aryl ring or the heteroaryl ring is joined to the remainder of the molecule.
  • heteroaryl includes 5-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen.
  • Example heteroaryl groups include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo[1,5-b]pyridazinyl, imidazol[1,2-a]pyrimidinyl and purinyl, as well as benzo-fused derivatives, for example benzoxazolyl, benzofuryl, benzothiazolyl, benzothiadiazol
  • Heteroaryl groups can be optionally substituted.
  • substituents for “optionally substituted heteroaryls” include one to four instances of F, Cl, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(O)CH 3 , COOH, CO 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, SO 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.
  • a heterocyclyl group is attached at a carbon atom of the heterocyclyl group.
  • carbon bonded heterocyclyl groups include bonding arrangements at position 2, 3, 4, 5, or 6 of a pyridine ring, position 3, 4, 5, or 6 of a pyridazine ring, position 2, 4, 5, or 6 of a pyrimidine ring, position 2, 3, 5, or 6 of a pyrazine ring, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole ring, position 2, 4, or 5 of an oxazole, imidazole or thiazole ring, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole ring, position 2 or 3 of an aziridine ring, position 2, 3, or 4 of an azetidine ring, position 2, 3, 4, 5, 6, 7, or 8 of a
  • the heterocyclyl group is N-attached.
  • nitrogen bonded heterocyclyl or heteroaryl groups include bonding arrangements at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H-indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or ⁇ -carboline.
  • alkoxy refers to a linear or branched monovalent radical represented by the formula —OR in which R is alkyl, as defined herein. Alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, mono-, di- and tri-fluoromethoxy and cyclopropoxy. “Haloalkoxy” refers to a haloalkyl group, as that term is defined herein, as R.
  • alkanoyl refers to group (alkyl)-C( ⁇ O)—, wherein alkyl is as defined herein.
  • C 1 -C 6 alkanoyl refers to a group of formula (C 1 -C 5 alkyl)-C( ⁇ O)—.
  • Alkanoyl groups include, formyl, acetyl, propanoyl, isopropanoyl, butanoyl, isobutanoyl, pentanoyl, 3-methylpentanoyl, and hexanoyl.
  • Optionally substituted unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 0, 1, 2, 3, 4, or 5 or more, or any range derivable therein) of the substituents listed for that group in which said substituents may be the same or different.
  • an optionally substituted group has 1 substituent.
  • an optionally substituted group has 2 substituents.
  • an optionally substituted group has 3 substituents.
  • an optionally substituted group has 4 substituents.
  • an optionally substituted group has 5 substituents.
  • a wavy line “ ” that intersects a bond in a chemical structure indicate the point of attachment of the atom to which the wavy bond is connected in the chemical structure to the remainder of a molecule, or to the remainder of a fragment of a molecule.
  • an arrow together with an asterisk is used in the manner of a wavy line to indicate a point of attachment.
  • divalent groups are described generically without specific bonding configurations. It is understood that the generic description is meant to include both bonding configurations, unless specified otherwise.
  • R 1 —R 2 —R 3 if the group R 2 is described as —CH 2 C(O)—, then it is understood that this group can be bonded both as R 1 —CH 2 C(O)—R 3 , and as R 1 —C(O)CH 2 —R 3 , unless specified otherwise.
  • phrases “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate.
  • Compounds of the invention may be in the form of a salt, such as a pharmaceutically acceptable salt.
  • “Pharmaceutically acceptable salts” include both acid and base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid and the like, and organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascor
  • “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Particular base addition salts are the ammonium, potassium, sodium, calcium and magnesium salts.
  • Salts derived from pharmaceutically acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particular organic non-toxic bases include isopropylamine, diethylamine, ethanolamine, tromethamine, dicyclohexylamine, choline, and caffeine.
  • a salt is selected from a hydrochloride, hydrobromide, trifluoroacetate, sulphate, phosphate, acetate, fumarate, maleate, tartrate, lactate, citrate, pyruvate, succinate, oxalate, methanesulphonate, p-toluenesulphonate, bisulphate, benzenesulphonate, ethanesulphonate, malonate, xinafoate, ascorbate, oleate, nicotinate, saccharinate, adipate, formate, glycolate, palmitate, L-lactate, D-lactate, aspartate, malate, L-tartrate, D-tartrate, stearate, furoate (e.g., 2-furoate or 3-furoate), napadisylate (naphthalene-1,5-disulfonate or naphthalene-1-(sulfonic acid)-5
  • a “sterile” formulation is aseptic or free from all living microorganisms and their spores.
  • Stereoisomers refer to compounds that have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers and the like.
  • Chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties or biological activities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • d and l or (+) and ( ⁇ ) are employed to designate the sign of rotation of plane-polarized light by the compound, with ( ⁇ ) or l meaning that the compound is levorotatory.
  • a compound prefixed with (+) or d is dextrorotatory.
  • these stereoisomers are identical except that they are mirror images of one another.
  • a specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.
  • tautomer or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • Certain compounds of the invention can exist in unsolvated forms as well as solvated forms, including hydrated forms.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the present invention. Examples of solvents that form solvates include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • Certain compounds of the invention can exist in multiple crystalline or amorphous forms. In general, all physical forms are intended to be within the scope of the present invention.
  • the term “hydrate” refers to the complex where the solvent molecule is water.
  • a “metabolite” refers to a product produced through metabolism in the body of a specified compound or salt thereof. Such products can result, for example, from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound.
  • Metabolite products typically are identified by preparing a radiolabelled (e.g., 14 C or 3 H) isotope of a compound of the invention, administering it in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples.
  • a detectable dose e.g., greater than about 0.5 mg/kg
  • an animal such as rat, mouse, guinea pig, monkey, or to a human
  • a detectable dose typically about 30 seconds to 30 hours
  • the metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis.
  • metabolites In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art.
  • the metabolite products so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.
  • amino-protecting group refers to a derivative of the groups commonly employed to block or protect an amino group while reactions are carried out on other functional groups on the compound.
  • protecting groups include carbamates, amides, alkyl and aryl groups, and imines, as well as many N-heteroatom derivatives which can be removed to regenerate the desired amine group.
  • Particular amino protecting groups are Pmb (p-Methoxybenzyl), Boc (tert-Butyloxycarbonyl), Fmoc (9-Fluorenylmethyloxycarbonyl) and Cbz (Carbobenzyloxy). Further examples of these groups are found in T. W. Greene and P. G. M.
  • protected amino refers to an amino group substituted with one of the above amino-protecting groups.
  • Carboxy-protecting group refers to those groups that are stable to the conditions of subsequent reaction(s) at other positions of the molecule, which may be removed at the appropriate point without disrupting the remainder of the molecule, to give the unprotected carboxy-group.
  • carboxy protecting groups include, ester groups and heterocyclyl groups. Ester derivatives of the carboxylic acid group may be employed to block or protect the carboxylic acid group while reactions are carried out on other functional groups on the compound.
  • ester groups include substituted arylalkyl, including substituted benzyls, such as 4-nitrobenzyl, 4-methoxybenzyl, 3,4-dimethoxybenzyl, 2,4-dimethoxybenzyl, 2,4,6-trimethoxybenzyl, 2,4,6-trimethylbenzyl, pentamethylbenzyl, 3,4-methylenedioxybenzyl, benzhydryl, 4,4′-dimethoxybenzhydryl, 2,2′,4,4′-tetramethoxybenzhydryl, alkyl or substituted alkyl esters such as methyl, ethyl, t-butyl allyl or t-amyl, triphenylmethyl (trityl), 4-methoxytrityl, 4,4′-dimethoxytrityl, 4,4′,4′′-trimethoxytrityl, 2-phenylprop-2-yl, thioesters such as t-butyl thioest
  • carboxy-protecting groups are heterocyclyl groups such as 1,3-oxazolinyl. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, “Protecting Groups in Organic Synthesis, 3 rd ed., John Wiley & Sons, Inc., 1999.
  • protected carboxy refers to a carboxy group substituted with one of the above carboxy-protecting groups.
  • “Hydroxy-protecting group” refers to a derivative of the hydroxy group commonly employed to block or protect the hydroxy group while reactions are carried out on other functional groups on the compound.
  • protecting groups include tetrahydropyranyloxy, benzoyl, acetoxy, carbamoyloxy, benzyl, and silylethers (e.g., TBS, TBDPS) groups. Further examples of these groups are found in T. W. Greene and P. G. M. Wuts, “Protecting Groups in Organic Synthesis, 3 rd ed., John Wiley & Sons, Inc., 1999.
  • the term “protected hydroxy” refers to a hydroxy group substituted with one of the above hydroxy-protecting groups.
  • Compounds of the invention may contain one or more asymmetric carbon atoms. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof.
  • the syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Mixtures of particular diastereomeric compounds may be separated, or enriched in one or more particular diastereomers, by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated, or enantiomerically enriched, using the same techniques or others known in the art.
  • Each of the asymmetric carbon or nitrogen atoms may be in the R or S configuration and both of these configurations are within the scope of the invention.
  • stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined. Unless otherwise specified, if solid wedges or dashed lines are used, relative stereochemistry is intended.
  • Another aspect includes prodrugs of the compounds of the invention including known amino-protecting and carboxy-protecting groups which are released, for example hydrolyzed, to yield the compound of the present invention under physiologic conditions.
  • prodrug refers to a precursor or derivative form of a pharmaceutically active substance that is less efficacious to the patient compared to the parent drug and is capable of being enzymatically or hydrolytically activated or converted into the more active parent form. See, e.g., Wilman, “Prodrugs in Cancer Chemotherapy” Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Harbor (1986) and Stella et al., “Prodrugs: A Chemical Approach to Targeted Drug Delivery,” Directed Drug Delivery, Borchardt et al., (ed.), pp. 247-267, Humana Press (1985).
  • Prodrugs include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, ⁇ -lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, and 5-fluorocytosine and 5-fluorouridine prodrugs.
  • a particular class of prodrugs are compounds in which a nitrogen atom in an amino, amidino, aminoalkyleneamino, iminoalkyleneamino or guanidino group is substituted with a hydroxy group, an alkylcarbonyl (—CO—R) group, an alkoxycarbonyl (—CO—OR), or an acyloxyalkyl-alkoxycarbonyl (—CO—O—R—O—CO—R) group where R is a monovalent or divalent group, for example alkyl, alkylene or aryl, or a group having the Formula —C(O)—O—CP1P2-haloalkyl, where P1 and P2 are the same or different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl or aryl.
  • the nitrogen atom is one of the nitrogen atoms of the amidino group of the compounds of the invention.
  • Prodrugs may be prepared by reacting a compound of the present invention with an activated group, such as acyl groups, to bond, for example, a nitrogen atom in the compound to the exemplary carbonyl of the activated acyl group.
  • activated carbonyl compounds are those containing a leaving group bonded to the carbonyl group, and include, for example, acyl halides, acyl amines, acyl pyridinium salts, acyl alkoxides, acyl phenoxides such as p-nitrophenoxy acyl, dinitrophenoxy acyl, fluorophenoxy acyl, and difluorophenoxy acyl.
  • the reactions are generally carried out in inert solvents at reduced temperatures such as ⁇ 78 to about 50° C.
  • the reactions may also be carried out in the presence of an inorganic base, for example potassium carbonate or sodium bicarbonate, or an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine, or the like.
  • prodrugs are also encompassed.
  • a free carboxyl group of a compound of the invention can be derivatized as an amide or alkyl ester.
  • compounds of the invention comprising free hydroxy groups can be derivatized as prodrugs by converting the hydroxy group into a group such as, but not limited to, a phosphate ester, hemisuccinate, dimethylaminoacetate, or phosphoryloxymethyloxycarbonyl group, as outlined in Fleisher, D. et al., (1996) Improved oral drug delivery: solubility limitations overcome by the use of prodrugs Advanced Drug Delivery Reviews, 19:115.
  • Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups.
  • Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group can be an alkyl ester optionally substituted with groups including, but not limited to, ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed.
  • Prodrugs of this type are described in J. Med. Chem., (1996), 39:10.
  • More specific examples include replacement of the hydrogen atom of the alcohol group with a group such as (C 1 -C 6 )alkanoyloxymethyl, 1-((C 1 -C 6 )alkanoyloxy)ethyl, 1-methyl-1-((C 1 -C 6 )alkanoyloxy)ethyl, (C 1 -C 6 )alkoxycarbonyloxymethyl, N—(C 1 -C 6 )alkoxycarbonylaminomethyl, succinoyl, (C 1 -C 6 )alkanoyl, alpha-amino(C 1 -C 4 )alkanoyl, arylacyl and alpha-aminoacyl, or alpha-aminoacyl-alpha-aminoacyl, where each alpha-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(OH) 2 , —P(O)(O(C 1 -C 6 )alky
  • leaving group refers to a portion of a first reactant in a chemical reaction that is displaced from the first reactant in the chemical reaction.
  • Examples of leaving groups include, but are not limited to, halogen atoms, alkoxy and sulfonyloxy groups.
  • Example sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trifluoromethylsulfonyloxy (triflate group)) and arylsulfonyloxy groups (for example p-toluenesulfonyloxy (tosylate group) and p-nitrosulfonyloxy (nosylate group)).
  • a “subject,” “individual,” or “patient” is a vertebrate.
  • the vertebrate is a mammal.
  • Mammals include, but are not limited to, farm animals (such as cows), sport animals, pets (such as guinea pigs, cats, dogs, rabbits and horses), primates, mice and rats.
  • a mammal is a human.
  • the patient is typically in need thereof.
  • inhibiting includes any measurable decrease or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, reduction of activity (e.g., IRAK4 activity) compared to normal.
  • IRAK4 activity e.g., IRAK4 activity
  • a compound of Formula 0, Formula I, or Formula II is selective for inhibition of IRAK4 over IRAK1.
  • selective for inhibition it is meant that the compound is at least a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, better inhibitor of IRAK4 activity compared to IRAK1 activity, or is at least a 2-, 3-, 4-, 5-, 10-, 25-, 50-, 100-, 250-, or 500-fold better inhibitor of IRAK4 activity compared to IRAK1 activity.
  • a “therapeutically effective amount” means an amount of a compound of the present invention, such as a compound of Formula 0, Formula I, or Formula II (e.g., a compound of Tables 1, 2 or 3), that (i) treats or prevents the particular disease, condition or disorder, or (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, and optionally (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein.
  • the therapeutically effective amount is an amount sufficient to decrease or alleviate the symptoms of an autoimmune or inflammatory disease (e.g., lupus).
  • a therapeutically effective amount is an amount of a chemical entity described herein sufficient to significantly decrease the activity or number of B-cells.
  • the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth or kill existing cancer cells, it may be cytostatic or cytotoxic.
  • efficacy can, for example, be measured by assessing the time to disease progression (TTP) or determining the response rate (RR).
  • Treatment refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, stabilized (i.e., not worsening) state of disease, decreasing the rate of disease progression, amelioration or palliation of the disease state, prolonging survival as compared to expected survival if not receiving treatment and remission or improved prognosis.
  • compounds of the invention are used to delay development of a disease or disorder or to slow the progression of a disease or disorder.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder, (for example, through a genetic mutation) or those in which the condition or disorder is to be prevented.
  • Inflammatory disorder refers to any disease, disorder or syndrome in which an excessive or unregulated inflammatory response leads to excessive inflammatory symptoms, host tissue damage, or loss of tissue function. “Inflammatory disorder” also refers to a pathological state mediated by influx of leukocytes or neutrophil chemotaxis.
  • Inflammation refers to a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute, or wall off (sequester) both the injurious agent and the injured tissue. Inflammation is notably associated with influx of leukocytes or neutrophil chemotaxis. Inflammation can result from infection with pathogenic organisms and viruses and from noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune responses to foreign antigens, and autoimmune responses.
  • inflammatory disorders amenable to treatment with a compound of the present invention encompass disorders associated with reactions of the specific defense system as well as with reactions of the nonspecific defense system.
  • Specific defense system refers to the component of the immune system that reacts to the presence of specific antigens. Examples of inflammation resulting from a response of the specific defense system include the classical response to foreign antigens, autoimmune diseases, and delayed type hypersensitivity responses mediated by T-cells. Chronic inflammatory diseases, the rejection of solid transplanted tissue and organs, e.g., kidney and bone marrow transplants, and graft versus host disease (GVHD), are further examples of inflammatory reactions of the specific defense system.
  • GVHD graft versus host disease
  • nonspecific defense system refers to inflammatory disorders that are mediated by leukocytes that are incapable of immunological memory (e.g., granulocytes, and macrophages).
  • inflammation that result, at least in part, from a reaction of the nonspecific defense system include inflammation associated with conditions such as adult (acute) respiratory distress syndrome (ARDS) or multiple organ injury syndromes; reperfusion injury; acute glomerulonephritis; reactive arthritis; dermatoses with acute inflammatory components; acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; thermal injury; inflammatory bowel disease; granulocyte transfusion associated syndromes; and cytokine-induced toxicity.
  • ARDS adult (acute) respiratory distress syndrome
  • multiple organ injury syndromes reperfusion injury
  • acute glomerulonephritis reactive arthritis
  • dermatoses with acute inflammatory components acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; thermal injury; inflammatory bowel disease; granulocyte transfusion associated syndromes; and
  • Autoimmune disease refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents.
  • Non-limiting examples of autoimmune diseases include rheumatoid arthritis, lupus and multiple sclerosis.
  • Allergic disease refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy.
  • Arthritic disease refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies.
  • Dermatis refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies.
  • Transplant rejection refers to any immune reaction directed against grafted tissue, such as organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia.
  • the therapeutic methods of the present invention include methods for the treatment of disorders associated with inflammatory cell activation.
  • “Inflammatory cell activation” refers to the induction by a stimulus (including, but not limited to, cytokines, antigens or auto-antibodies) of a proliferative cellular response, the production of soluble mediators (including but not limited to cytokines, oxygen radicals, enzymes, prostanoids, or vasoactive amines), or cell surface expression of new or increased numbers of mediators (including, but not limited to, major histocompatability antigens or cell adhesion molecules) in inflammatory cells (including but not limited to monocytes, macrophages, T lymphocytes, B lymphocytes, granulocytes (i.e., polymorphonuclear leukocytes such as neutrophils, basophils, and eosinophils), mast cells, dendritic cells, Langerhans cells, and endothelial cells). It will be appreciated by persons skilled in the art that the activation of one or a combination of these phenotypes in these cells can contribute to the initiation
  • inflammatory disorders which can be treated according to the methods of this invention include, but are not limited to, asthma, rhinitis (e.g., allergic rhinitis), allergic airway syndrome, atopic dermatitis, bronchitis, rheumatoid arthritis, psoriasis, lupus, chronic obstructive pulmonary disease (COPD), contact dermatitis, chronic obstructive pulmonary disease and delayed hypersensitivity reactions.
  • rhinitis e.g., allergic rhinitis
  • allergic airway syndrome e.g., atopic dermatitis, bronchitis, rheumatoid arthritis, psoriasis, lupus, chronic obstructive pulmonary disease (COPD), contact dermatitis, chronic obstructive pulmonary disease and delayed hypersensitivity reactions.
  • COPD chronic obstructive pulmonary disease
  • cancer and “cancerous”, “neoplasm”, and “tumor” and related terms refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include carcinoma, blastoma, sarcoma, seminoma, glioblastoma, melanoma, leukemia, and myeloid or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer) and lung cancer including small-cell lung cancer, non-small cell lung cancer (“NSCLC”), adenocarcinoma of the lung and squamous carcinoma of the lung.
  • NSCLC non-small cell lung cancer
  • cancers include skin, keratoacanthoma, follicular carcinoma, hairy cell leukemia, buccal cavity, pharynx (oral), lip, tongue, mouth, salivary gland, esophageal, larynx, hepatocellular, gastric, stomach, gastrointestinal, small intestine, large intestine, pancreatic, cervical, ovarian, liver, bladder, hepatoma, breast, colon, rectal, colorectal, genitourinary, biliary passage, thyroid, papillary, hepatic, endometrial, uterine, salivary gland, kidney or renal, prostate, testis, vulval, peritoneum, anal, penile, bone, multiple myeloma, B-cell lymphoma, diffuse large B-Cell lymphoma (DLBCL), central nervous system, brain, head and neck, Hodgkin's, and associated metastases.
  • DLBCL diffuse large B-Cell lymphoma
  • neoplastic disorders include myeloproliferative disorders, such as polycythemia vera, essential thrombocytosis, myelofibrosis, such as primary myelofibrosis, and chronic myelogenous leukemia (CML).
  • myeloproliferative disorders such as polycythemia vera, essential thrombocytosis, myelofibrosis, such as primary myelofibrosis, and chronic myelogenous leukemia (CML).
  • myeloproliferative disorders such as polycythemia vera, essential thrombocytosis, myelofibrosis, such as primary myelofibrosis, and chronic myelogenous leukemia (CML).
  • CML chronic myelogenous leukemia
  • chemotherapeutic agent is an agent useful in the treatment of a given disorder, for example, cancer or inflammatory disorders.
  • chemotherapeutic agents are well-known in the art and include examples such as those disclosed in U.S. Publ. Appl. No. 2010/0048557, incorporated herein by reference. Additionally, chemotherapeutic agents include pharmaceutically acceptable salts, acids or derivatives of any of chemotherapeutic agents, as well as combinations of two or more of them.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 Cl, 123 I, and 125 I, respectively.
  • Isotopically-labeled compounds e.g., those labeled with 3 H and 14 C
  • Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements).
  • one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon.
  • Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • Isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed in the Schemes or in the Examples herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention.
  • any compound or composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any compound or composition of the invention.
  • a or “an” means one or more, unless clearly indicated otherwise.
  • another means at least a second or more.
  • one aspect of the invention includes a compound of Formula 0:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 12 NR 6 R 7 , —NR 8 R 9 , —O—C 1-3 alkyl, a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group, heterocyclic group, heteroaryl ring, or aryl ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 1-2 NR 10 R 11 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, —C(O)NR 8 R 9 , or —NR 8 R 9 ,
  • any alkyl, alkenyl, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-7 membered monocyclic saturated or partially saturated heterocyclic group that may be optionally substituted with oxo;
  • R 5 is hydrogen, —CN, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —C(O)NR 8 R 9 , —O(C 3-7 cycloalkyl group), —O(C 1-3 alkyl)-3-8 membered cycloalkyl group, —O(C 0-3 alkyl)-3-8 membered saturated or partially saturated heterocyclic group, —O(C 1-3 alkyl)-phenyl, a —O(C 1-3 alkyl)-5-6 membered heteroaryl ring, a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-11 membered saturated or partially saturated heterocyclic group that may be optionally substituted with (i) —C(O)(C 1-3 alkyl) optionally substituted with halogen or (ii) with C 1-3 alkyl optionally substituted with halogen, and
  • any cycloalkyl group, heterocyclic group, phenyl, or heteroaryl ring is optionally substituted by halogen; oxo; CN; OH; C 1-6 alkoxy; —NR 8 R 9 ; —C(O)(C 1-3 alkyl); —(C 0-3 alkyl)C(O)NR 10 R 11 ; —S(O) 12 NR 8 R 9 ; —OP(O)(OC 1-3 alkyl) 2 ; C 3-10 cycloalkyl group optionally substituted with OH or halogen; a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted with oxo or C 1-3 alkyl; a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl; or C 1-4 alkyl optionally substituted by halogen, oxo
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, —(C 0-3 alkyl)-C 3-6 cycloalkyl group, a —(C 0-3 alkyl)-3-11 membered heterocyclic group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 1-2 R 13 , —C(O)(C 1-3 alkyl), —C(O)NR 10 R 11 , —C(O)OR 13 , —S(O) 1-2 N 10 R 11 , or —(C 0-3 alkyl)-OP(O)(OC 1-3 alkyl) 2 ,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen; oxo; CN; OR 13 ; C 1-3 haloalkoxy; —C(O)(C 1-3 alkyl); —S—C 1-3 alkyl; or C 1-3 alkyl optionally substituted with OH, halogen, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-8 membered heterocyclic group, and
  • A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, —(C 0-3 alkyl)-phenyl, a 3-11 membered saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 12 R 13 , or R 10 and R 11 are taken together to form a 5-8 membered heterocyclic group,
  • any alkyl, cycloalkyl group, phenyl, heterocyclic group, or heteroaryl ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , —NR 6 R 7 , or a 5-6 membered monocyclic heteroaryl ring;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is hydrogen, halogen, CN, or C 1-3 alkyl optionally substituted with —NH 2 , halogen, or CN.
  • Another aspect of the invention includes a compound of Formula I:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 1-2 NR 6 R 7 , —NR 8 R 9 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 12 NR 10 R 11 , or —NR 8 R 9 ;
  • any alkyl or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —O(C 3-7 cycloalkyl group), —O(C 1-3 alkyl)-3-8 membered cycloalkyl group, —O(C 1-3 alkyl)-3-8 membered saturated or partially saturated heterocyclic group, —O(C 1-3 alkyl)-phenyl, a —O(C 1-3 alkyl)-5-6 membered heteroaryl ring, a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is optionally substituted by halogen, oxo, CN, OH, C 1-6 alkoxy, —C(O)(C 1-3 alkyl), —(C 0-3 alkyl)C(O)NR 10 R 11 , —S(O) 1-2 NR 8 R 9 , —OP(O)(OC 1-3 alkyl) 2 , a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl, or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, phenyl, a 3-8 membered saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or —NR 8 R 9 ;
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, —(C 0-3 alkyl)-C 3-6 cycloalkyl group, a —(C 0 -C 3 alkyl)-3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)(C 1-3 alkyl), —C(O)NR 10 R 11 , —C(O)OR 13 , —S(O) 12 NR 10 R 11 , or —OP(O)(OC 1-3 alkyl) 2 ,
  • any alkyl, cycloalkyl group, or heterocyclic group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, or a 3-8 membered heterocyclic group;
  • A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, —(C 0-3 alkyl)-phenyl, a 3-11 membered saturated heterocyclic group, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 1-2 R 13 , or R 10 and R 11 are taken together to form a 5-8 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , or —NR 6 R 7 ;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is H, —Cl, —CN, or —CH 3 .
  • Another aspect of the invention includes a compound of Formula II:
  • R 1 is hydrogen or halogen
  • R 3 is hydrogen, halogen, CN, OH, C 1-3 alkyl, C 2-3 alkenyl, C 3-7 cycloalkyl group, C 1 -C 3 alkanoyl, —(C 0 -C 3 alkyl)C(O)NR 6 R 7 , —(C 2-3 alkenyl)C(O)NR 6 R 7 , —S(O) 12 NR 6 R 7 , —NR 8 R 9 , a 3-7 membered monocyclic saturated or partially saturated heterocyclic group, a 5-6 membered monocyclic heteroaryl ring, or a 5-6 membered monocyclic aryl ring,
  • any alkyl, alkanoyl, or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl;
  • R 4 is hydrogen, halogen, C 1-3 alkyl, C 2-3 alkenyl, —(C 0 -C 3 alkyl)C(O)R 13 —(C 2-3 alkenyl)C(O)NR 10 R 11 , —S(O) 12 NR 10 R 11 , or —NR 8 R 9 ;
  • any alkyl or alkenyl is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 5 is hydrogen, C 1-6 alkyl, C 1-6 alkoxy, C 3-10 cycloalkyl group, —NR 8 R 9 , —O(C 3-7 cycloalkyl group), a 3-11 membered saturated or partially saturated heterocyclic group, or a 5-6 membered monocyclic heteroaryl ring,
  • any alkyl or alkoxy is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkoxy, or C 1-3 haloalkoxy, and
  • any cycloalkyl group or other ring is optionally substituted by halogen, oxo, CN, OH, —(C 0 -C 3 alkyl)C(O)NR 10 R 11 , —OP(O)(OC 1-3 alkyl) 2 , or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, or —NR 8 R 9 ;
  • A is a 3-11 membered heterocyclic group optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , or —S(O) 1-2 NR 10 R 11
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • A is a 5-membered nitrogen containing heterocyclic group, the nitrogen atom is substituted;
  • R 6 and R 7 are, independently at each occurrence, hydrogen, C 1-3 alkyl, or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 8 , R 9 , R 10 and R 11 are, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl group, a 3-11 membered saturated heterocyclic group, —C(O)R 13 , —C(O)OR 13 , —C(O)NR 6 R 7 , or —S(O) 1-2 R 13
  • any alkyl, cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 13 , or —NR 6 R 7 ;
  • R 12 is, independently at each occurrence, hydrogen, C 1-6 alkyl or C 3-6 cycloalkyl group,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy;
  • R 13 is, independently at each occurrence, hydrogen, C 1-6 alkyl, C 3-10 cycloalkyl group, or a 3-11 membered saturated heterocyclic group,
  • any alkyl, cycloalkyl group, or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, C 1-3 haloalkoxy, —OR 12 , or —NR 6 R 7 ; and
  • R 16 is H, —Cl, —CN, or —CH 3 .
  • R 1 and R 4 are each hydrogen, and R 3 is hydrogen, OH, halogen, CH 3 , CH 2 OH, CH 2 F, OCHF 2 , CHF 2 , CF 3 , cyclopropyl, azetidinyl, CN, —C(O)CH 3 , —C(O)NH 2 , —C(O)NHCH 3 , —NHCH 3 , —SO 2 —NH 2 , or —SO 2 —NHCH 3 .
  • R 1 and R 4 are each hydrogen, and R 3 is hydrogen, halogen, CH 3 , CH 2 F, CHF 2 , CF 3 , cyclopropyl, or —C(O)CH 3 .
  • R 1 and R 4 are each hydrogen, and R 3 is hydrogen, OH, CH 3 , or CH 2 OH.
  • R 1 and R 4 are each hydrogen, and R 3 is Br, Cl, F, OCHF 2 , CHF 2 , or CF 3 .
  • R 1 and R 4 are each hydrogen, and R 3 is cyclopropyl, azetidinyl, CN, —C(O)CH 3 , —C(O)NH 2 , —C(O)NHCH 3 , —NHCH 3 , —SO 2 —NH 2 , or —SO 2 —NHCH 3 .
  • R 1 and R 3 are each hydrogen, and R 4 is Cl, CHF 2 ,
  • R 1 and R 3 are each hydrogen, and R 4 is hydrogen or CH 3 .
  • R 1 , R 4 and R 3 are each hydrogen.
  • R 16 is H. In some embodiments, R 16 is —Cl, —CN, or —CH 3 .
  • R 5 is a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted by halogen; oxo; CN; OH; C 1-6 alkoxy; —NR 8 R 9 ; —C(O)(C 1-3 alkyl); —(C 0 -C 3 alkyl)C(O)NR 10 R 11 ; —S(O) 12 NR 8 R 9 ; —OP(O)(OC 1-3 alkyl) 2 ; C 3-10 cycloalkyl group optionally substituted with OH or halogen; a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted with oxo or C 1-3 alkyl; a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl; or C 1-4 alkyl optionally substituted by halogen, oxo, CN,
  • R 5 is a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted by halogen, oxo, CN, OH, —(C 0-3 alkyl)C(O)NR 10 R 11 , —OP(O)(OC 1-3 alkyl) 2 , or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, or —NR 8 R 9 .
  • R 5 is an N-linked 3-11 membered saturated heterocyclic group optionally substituted by halogen, oxo, CN, OH, —(C 0-3 alkyl)C(O)NR 10 R 11 , —OP(O)(OC 1-3 alkyl) 2 , or C 1-3 alkyl optionally substituted by halogen, oxo, CN, OH, or —NR 8 R 9 .
  • the ring heteroatoms of the 3-11 membered saturated or partially saturated heterocyclic group of R 5 are selected from nitrogen and oxygen.
  • R 5 is piperidinyl, piperazinyl, or morpholinyl, wherein any R 5 is optionally substituted by halogen; oxo; CN; OH; C 1-6 alkoxy; —NR 8 R 9 ; —C(O)(C 1-3 alkyl); —(C 0-3 alkyl)C(O)NR 10 R 11 ; —S(O) 12 NR 8 R 9 ; —OP(O)(OC 1-3 alkyl) 2 ; C 3-10 cycloalkyl group optionally substituted with OH or halogen; a 3-11 membered saturated or partially saturated heterocyclic group optionally substituted with oxo or C 1-3 alkyl; a 5-6 membered monocyclic heteroaryl ring optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, or C 1-3 haloalkyl; or C 1-4 alkyl optionally substituted by halogen, o
  • R 5 is N-linked piperidinyl, N-linked piperazinyl, or N-linked morpholinyl, wherein any R 5 is optionally substituted by halogen, oxo, CN, OH, or C 1-3 alkyl optionally substituted by halogen, oxo, CN, or OH.
  • R 5 is:
  • R 5 is:
  • R 5 is
  • A is a 5-membered nitrogen containing heterocyclic ring
  • the nitrogen atom is substituted.
  • A comprises one or more nitrogen atoms, e.g., 2, 3, or more nitrogen atoms
  • each nitrogen atom is substituted. More particularly, in some embodiments, the compound is not
  • A is a 3-11 membered, non-aromatic heterocyclic group.
  • A is a 3-11 membered heterocyclic group comprising at least one oxygen as a ring atom and is optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , or —S(O) 12 NR 10 R 11 , wherein any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy.
  • A is a 5 or 6 membered ring optionally containing an additional ring heteroatom
  • A is optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , or —S(O) 1-2 NR 10 R 11 ,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy.
  • 0-A, I-A, or II-A is further defined as 0-B, I-B, or II-B:
  • R 14 and R 15 are each selected from halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , and —S(O) 1-2 NR 10 R 11 ,
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy; or R 14 and R 15 together form a C 3-6 cycloalkyl group or saturated or partially saturated 3-6 membered heterocyclic group,
  • any cycloalkyl group or other ring is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy.
  • A does not contain oxygen as a ring atom and is optionally substituted by halogen, oxo, CN, OH, C 1-6 alkyl, C 3-6 cycloalkyl group, —NR 8 R 9 , —NR 12 C(O)R 13 , —NR 12 S(O) 12 R 13 , —C(O)NR 10 R 11 , —C(O)OR 13 , or —S(O) 1-2 NR 10 R 11 wherein any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy.
  • any alkyl or cycloalkyl group is independently optionally substituted by halogen, oxo, CN, OH, C 1-3 alkyl, C 1-3 haloalkyl, C 1-3 alkoxy, or C 1-3 haloalkoxy.
  • a compound is selected from the group consisting of the compounds of Tables 1, 2 and 3, shown below, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • reaction Schemes below provide routes for synthesizing the compounds of the invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used. Although some specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be substituted to provide a variety of derivatives or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • the starting materials and the intermediates of the synthetic reaction schemes can be isolated and purified if desired using conventional techniques, including but not limited to, filtration, distillation, crystallization, chromatography, and the like. Such materials can be characterized using conventional means, including physical constants and spectral data.
  • the reactions described herein preferably are conducted under an inert atmosphere at atmospheric pressure at a reaction temperature range of from about ⁇ 78° C. to about 150° C., more preferably from about 0° C. to about 125° C., and most preferably and conveniently at about room (or ambient) temperature, or, about 20° C.
  • requisite 3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl derivatives can be prepared by nitration of 7-halo-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl derivatives I-la to afford I-1b followed by displacement of the halogen with an amine to afford I-2a.
  • Typical amines include morpholine and 3-hydroxymethyl piperidine.
  • the requisite precursors are commercially available or are prepared as described herein.
  • Aromatic nitration is well known and can be conducted under a variety of conditions known in the art. Nitration can be carried out, for example, by exposing an aromatic compound to concentrated nitric acid and sulfuric acid. Active substrates can be nitrated with HNO 3 alone or in H 2 O, HOAc and acetic anhydride and active compounds may be oxidized by mixtures of HNO 3 and H 2 SO 4 . Other nitrating reagents include NaNO 3 /TFA, Cu(NO 3 ) 2 /HOAc/Ac 2 O, N 2 O 4 , NO 2 + BF 4 ⁇ , NO 2 +PF 6 ⁇ and NO 2 + CF 3 SO 4 ⁇ . See, e.g., J. March, Advanced Organic Chemistry, John Wiley & Sons: New York, N.Y., 1992, pp. 522-23.
  • Reduction of the nitro group can be carried out with a variety of well-known reducing agents.
  • the nitro can be reduced under a hydrogen atmosphere in the presence of an inert solvent and in the presence of a metal effective to catalyze hydrogenation reactions such as platinum or palladium.
  • the reduction can also be carried out with an activated metal such as activated iron, zinc or tin (produced for example by washing iron powder with a dilute acid solution such as dilute hydrochloric acid).
  • Coupling of the amine 2b intermediate with 3 is achieved with commonly used coupling reagents or, alternatively, 3 can be converted to the corresponding acid chloride and condensed with 2b.
  • Acylation of a primary amine with an acid chloride is typically carried out in an inert solvent such as DMF, DCM, THF, pyridine with or without water as a co-solvent, at temperatures between 0° C. and 60° C. generally in the presence of a base such as Na 2 CO 3 , NaHCO 3 , K 2 CO 3 , DIPEA, TEA or pyridine and the like to afford the corresponding amide.
  • an inert solvent such as DMF, DCM, THF, pyridine with or without water as a co-solvent
  • Carboxylic acids can be converted into their acid chlorides using standard reagents well known to someone skilled in the art, such as thionyl chloride, oxalyl chloride, phosphoryl chloride and the like. Those reagents can be used in presence of bases such as DIPEA, TEA or pyridine.
  • a carboxylic acid can be converted in situ into activated derivatives by utilizing reagents developed for peptide synthesis which are well known to those skilled in the art. These activated acids were reacted directly with the amines as described to afford the corresponding amide.
  • Common coupling reagents include EDC, DCC, benzotriazol-1-yloxy-tri s(dimethylamino)-phosphonium hexafluorophosphate (BOP), bromo-tris-pyrrolidinophosphonium hexafluorophosphate (PyBrOP), 2-fluoro-1-methylpyridinium p-toluenesulphonate (Mukaiyama's reagent), O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU), 1-hydroxy-7-azabenzotriazole (HOAt) or (3-hydroxy-3H-1,2,3-triazolo[4,5-b]pyridinato-O)tri-1-pyrrolidinylphosphonium hexafluorophosphate (PyAOP) optionally in the presence of modifiers such as HOBt, with or without
  • 5,6-Diamino-2,2-dimethyl-3H-benzofuran derivatives exemplified herein can be prepared utilizing a 2,2-dimethyl-5-nitro-6-halo-3H-benzofuran II-3b as the key intermediate as depicted in Scheme II.
  • Addition of a methyl Grignard to methyl 5-chloro-2-fluoro-phenylacetate affords the tertiary alcohol II-2 which undergoes an intra-molecular cyclization to afford the II-3a.
  • Nitration and displacement of the chloride with an amine followed by reduction of the nitro and acylation with I-3 is carried out in analogy with Scheme I.
  • One skilled in the art will appreciate that the corresponding 5-fluoro and 5-bromo derivatives are readily available from methyl 2,5-difluorophenylacetate and methyl 5-bromo-2-fluorophenylacetate, respectively.
  • ether substituents can be readily prepared by reacting 5-amino-2,2-dimethyl-6-fluoro-3H-benzofuran (III-2b) with a alcohol in the presence of potassium tert-butoxide or other suitable strong base. Subsequent reduction of the nitro group, acylation of the resulting amine and any subsequent deprotection which may be required are carried out using standard methodology.
  • 3H-benzothiophene-1,1-dioxide derivatives can be prepared by intramolecular cyclization methyl 2-methylsulfonyl-4-morpholino-5-nitro-benzoate (V-3b) to afford morpholino-5-nitro-1,1-dioxo-benzothiophen-3-one (V-4) which is subsequently reduced to V-6b utilizing a two-step sequence comprising sodium borohydride reduction followed by triethylsilane/TFA reduction that is carried on as previously described. Reduction of the nitro group and acylation of the resulting amine is carried out as described previously.
  • Compounds of the present invention are useful as IRAK4 inhibitors. Accordingly, in one embodiment is provided a method of contacting a cell, such as an ex vivo cell, with a compound of the present invention to inhibit IRAK4 activity in the cell.
  • compositions comprising a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient, carrier or diluent.
  • a pharmaceutically acceptable excipient carrier or diluent.
  • a method of preventing, treating, or lessening the severity of a disease or condition responsive to the inhibition of IRAK4 in a patient comprising administering to the patient a therapeutically effective amount of a compound of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • the disease is selected from the group consisting of Crohn's disease, ulcerative colitis, Irritable Bowel Disorder (IBD), asthma, graft versus host disease, allograft rejection, chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, systemic lupus erythematosus, lupus nephritis, cutaneous lupus, psoriasis, systemic onset juvenile idiopathic arthritis, multiple sclerosis, neuropathic pain, gout, and gouty arthritis.
  • IBD Irritable Bowel Disorder
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • other diseases and conditions responsive to the inhibition of IRAK4 that can be treated using a compound of the present invention include metabolic syndromes, atherosclerosis, and neurodegeneration.
  • a compound of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof in therapy.
  • use of a compound of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof is provided in the treatment of an inflammatory disease.
  • use of a compound of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof is provided for the preparation of a medicament for the treatment of an inflammatory disease.
  • a compound of the present invention, or a stereoisomer or pharmaceutically acceptable salt thereof is provided for use in the treatment of an inflammatory disease.
  • the present invention provides pharmaceutical compositions or medicaments containing the compounds of the invention and at least one therapeutically inert carrier, diluent or excipient, as well as methods of using the compounds of the invention to prepare such compositions and medicaments.
  • compounds of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof, with the desired degree of purity may be formulated by mixing with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a dosage form at ambient temperature and at the appropriate pH.
  • physiologically acceptable carriers i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a dosage form at ambient temperature and at the appropriate pH.
  • the pH of the formulation depends mainly on the particular use and the concentration of compound, but typically ranges anywhere from about 3 to about 8.
  • a compound of Formula 0, Formula I, or Formula II is formulated in an acetate buffer, at pH 5.
  • the compounds of Formula 0, Formula I, or Formula II are sterile.
  • the compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.
  • compositions are formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular disorder being treated, the severity of the disorder, the particular patient being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the “effective amount” of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to inhibit IRAK4 activity. Typically such amount may be below the amount that is toxic to normal cells, or the patient as a whole.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • polyesters for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)
  • polylactides copolymers of L-glutamic acid and gamma-ethyl-L-glutamate
  • non-degradable ethylene-vinyl acetate non-degradable ethylene-viny
  • a dose to treat human patients may range from about 0.1 mg to about 1000 mg of a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • a typical dose may be about 1 mg to about 300 mg of the compound.
  • a dose may be administered once a day (QD), twice per day (BID), or more frequently, depending on the pharmacokinetic and pharmacodynamic properties, including absorption, distribution, metabolism, and excretion of the particular compound.
  • toxicity factors may influence the dosage and administration regimen.
  • the pill, capsule, or tablet may be ingested daily or less frequently for a specified period of time. The regimen may be repeated for a number of cycles of therapy.
  • the compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, epidural and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration.
  • the compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc.
  • Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, H. C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems . Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy . Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, R. C., Handbook of Pharmaceutical Excipients , Chicago, Pharmaceutical Press, 2005.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing
  • tablets containing various excipients such as citric acid may be employed together with various disintegrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia.
  • disintegrants such as starch, alginic acid and certain complex silicates
  • binding agents such as sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often useful for tableting purposes.
  • Solid compositions of a similar type may also be employed in soft and hard filled gelatin capsules. Preferred materials, therefore, include lactose or milk sugar and high molecular weight polyethylene glycols.
  • the active compound therein may be combined with various sweetening or flavoring agents, coloring matters or dyes and, if desired, emulsifying agents or suspending agents, together with diluents such as water, ethanol, propylene glycol, glycerin, or combinations thereof.
  • An example of a suitable oral dosage form is a tablet containing about 25 mg, 50 mg, 100 mg, 250 mg or 500 mg of the compound of the invention compounded with about 90-30 mg anhydrous lactose, about 5-40 mg sodium croscarmellose, about 5-30 mg polyvinylpyrrolidone (PVP) K30, and about 1-10 mg magnesium stearate.
  • the powdered ingredients are first mixed together and then mixed with a solution of the PVP.
  • the resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment.
  • An example of an aerosol formulation can be prepared by dissolving the compound, for example 5-400 mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such as sodium chloride, if desired.
  • the solution may be filtered, e.g., using a 0.2 micron filter, to remove impurities and contaminants.
  • the pharmaceutical composition also includes at least one additional anti-proliferative agent.
  • An embodiment therefore, includes a pharmaceutical composition comprising a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • a further embodiment includes a pharmaceutical composition comprising a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or excipient.
  • the invention further provides veterinary compositions comprising at least one active ingredient as above defined together with a veterinary carrier therefore.
  • Veterinary carriers are materials useful for the purpose of administering the composition and may be solid, liquid or gaseous materials which are otherwise inert or acceptable in the veterinary art and are compatible with the active ingredient. These veterinary compositions may be administered parenterally, orally or by any other desired route.
  • the compounds of Formula 0, Formula I, or Formula II may be employed alone or in combination with other therapeutic agents for the treatment of a disease or disorder described herein.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound of Formula 0, Formula I, or Formula II such that they do not adversely affect each other.
  • the combination therapy may provide “synergy” and prove “synergistic”, i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • the combination therapy may be administered as a simultaneous or sequential regimen.
  • the combination may be administered in two or more administrations.
  • the combined administration includes co-administration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • Combination therapies thus comprise the administration of at least one compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof, and the use of at least one other treatment method.
  • the amounts of the compound(s) of Formula 0, Formula I, or Formula II and the other pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • kits containing materials useful for the treatment of the diseases and disorders described above.
  • the kit comprises a container comprising a compound of Formula 0, Formula I, or Formula II, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • the kit may further comprise a label or package insert on or associated with the container.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • Suitable containers include, for example, bottles, vials, syringes, blister pack, etc.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container may hold a compound of Formula 0, Formula I, or Formula II or a formulation thereof which is effective for treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a compound of Formula 0, Formula I, or Formula II.
  • the article of manufacture may further comprise a second container comprising a pharmaceutical diluent, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • kits are suitable for the delivery of solid oral forms of a compound of Formula 0, Formula I, or Formula II, such as tablets or capsules.
  • a kit can include a number of unit dosages.
  • An example of such a kit is a “blister pack”. Blister packs are well known in the packaging industry and are widely used for packaging pharmaceutical unit dosage forms.
  • Step B 1-(4-Chloro-2-fluoro-phenyl)-2-methyl-propan-2-ol
  • Step B (1-(6-Amino-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)piperidin-4-yl)methanol
  • Step C N-(7-(4-(Hydroxymethyl)piperidin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B 6-Amino-7-(4-(hydroxymethyl)piperidin-1-yl)-2H-benzo[b][1,4]oxazin-3(4H)-one
  • Step C N-(7-(4-(Hydroxymethyl)piperidin-1-yl)-3-oxo-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step K Trans-2-trimethylsilylethyl N-[[5-(5-amino-2,2-dimethyl-3H-benzofuran-6-yl)-1,3-dioxan-2-yl]methyl]-N-methyl-carbamate
  • Step L Trans-2-trimethylsilylethyl N-[[5-[2,2-dimethyl-5-(pyrazolo[1,5-a]pyrimidine-3-carbonylamino)-3H-benzofuran-6-yl]-1,3-dioxan-2-yl]methyl]-N-methyl-carbamate
  • Step B (1-(5-Amino-2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)piperidin-4-yl)methanol
  • Step B 7-Fluoro-2,2-dimethyl-6-nitro-2H-benzo[b][1,4]oxazin-3(4H)-one
  • Step F N-(2,2-Dimethyl-7-morpholino-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • the mixture was purified by preparative HPLC (phenomenex, Gemini C18, 21.2 ⁇ 100 mm. 5 um, 110 A, A: acetonitrile 30-40%; B: 0.05% formic acid in water) to afford N-(2,2-dimethyl-7-morpholino-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (70 mg, 41%) as a yellow solid.
  • Step B (5-Amino-2-methyl-6-morpholino-2,3-dihydrobenzofuran-2-yl)methanol
  • Step B (1-(6-Nitro-3,4-dihydro-2H-benzo[b][1,4]oxazin-7-yl)piperidin-4-yl)methanol
  • Step D N-(7-(4-(Hydroxymethyl)piperidin-1-yl)-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)-6-methylpyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B 6-[4-(2,2-Difluoroethyl)piperazin-1-yl]-2,2-dimethyl-3H-benzofuran-5-amine
  • Step B 7-Morpholino-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-amine
  • Step B 4-Benzyl-7-fluoro-3-methyl-3,4-dihydro-2H-benzo[b][1,4]oxazine
  • Step F N-(3-Methyl-7-morpholino-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B 7-Chloro-2-methyl-6-nitro-2H-benzo[b][1,4]oxazin-3(4H)-one
  • Step F N-(2-Methyl-7-morpholino-3,4-dihydro-2H-benzo[b][1,4]oxazin-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B (S)-(1-(5-Amino-2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)pyrrolidin-3-yl)methanol
  • Step B (R)-(1-(5-Amino-2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)pyrrolidin-3-yl)methanol
  • Step C (R)—N-(6-(3-(Hydroxymethyl)pyrrolidin-1-yl)-2,2-dimethyl-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B (1-(5-Amino-2-(hydroxymethyl)-2-methyl-2,3-dihydrobenzofuran-6-yl)piperidin-4-yl)methanol
  • Step C N-(2-(Hydroxymethyl)-6-(4-(hydroxymethyl)piperidin-1-yl)-2-methyl-2,3-dihydrobenzofuran-5-yl)-6-methylpyrazolo[1,5-a]pyrimidine-3-carboxamide
  • the mixture was purified by preparative HPLC (Xbridge 21.2*250 mm c18, 10 um; A: acetonitrile 25-55%; B: 10 mM ammonium bicarbonate in water) to afford N-(2-(hydroxymethyl)-6-(4-(hydroxymethyl)piperidin-1-yl)-2-methyl-2,3-dihydrobenzofuran-5-yl)-6-methylpyrazolo[1,5-a]pyrimidine-3-carboxamide (65 mg, 53%) as a yellow solid.
  • Step B (5-Amino-6-(2,2-dimethylmorpholino)-2-methyl-2,3-dihydrobenzofuran-2-yl)methanol
  • Step C N-(6-(2,2-Dimethylmorpholino)-2-(hydroxymethyl)-2-methyl-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • the product was further purified by preparative HPLC ((Xbridge 21.2*250 mm c18, 10 um; A: acetonitrile 15-30%; B: 10 mM ammonium bicarbonate in water)) to afford N-(6-(2,2-dimethylmorpholino)-2-(hydroxymethyl)-2-methyl-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (48 mg, 58%) as a yellow solid.
  • Step B (1-(5-Amino-2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)-3-methylpyrrolidin-3-yl)methanol
  • Step C N-(6-(3-(Hydroxymethyl)-3-methylpyrrolidin-1-yl)-2,2-dimethyl-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B Cis-2-trimethylsilylethyl-N-[[5-(2,2-dimethyl-5-nitro-3H-benzofuran-6-yl)-1,3-dioxan-2-yl]methyl]-N-methyl-carbamate
  • Step D Cis-2-trimethylsilylethyl-N-[[5-[2,2-dimethyl-5-(pyrazolo[1,5-a]pyrimidine-3-carbonylamino)-3H-benzofuran-6-yl]-1,3-dioxan-2-yl]methyl]-N-methyl-carbamate
  • Step F N-(2-Ethyl-2-methyl-6-morpholino-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step F N-(2-Cyclopropyl-2-methyl-6-morpholino-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step F 5-Amino-N,N, 2-trimethyl-6-morpholino-2,3-dihydrobenzofuran-2-carboxamide
  • Step G N-(2-(Dimethylcarbamoyl)-2-methyl-6-morpholino-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B tert-Butyl 4-(2,2,2-trifluoro-1-(4-methoxybenzylamino)ethyl)piperidine-1-carboxylate
  • Step C 2,2,2-Trifluoro-N-[(4-methoxyphenyl)methyl]-1-(4-piperidyl)ethanamine trifluoroacetic acid salt
  • Step D 1-[1-(2,2-Dimethyl-5-nitro-3H-benzofuran-6-yl)-4-piperidyl]-2,2,2-trifluoro-N-[(4-methoxyphenyl)methyl]ethanamine
  • Step E tert-Butyl 4-[(4-amino-3-pyridyl)oxy]piperidine-1-carboxylate
  • Step F N-(2,2-Dimethyl-6-(4-(2,2,2-trifluoro-1-((4-methoxybenzyl)amino)ethyl)piperidin-1-yl)-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step G N-[6-[4-(1-Amino-2,2,2-trifluoro-ethyl)-1-piperidyl]-2,2-dimethyl-3H-benzofuran-5-yl]pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step B 4-(2-(Methoxymethyl)-2-methyl-5-nitro-2,3-dihydrobenzofuran-6-yl)morpholine
  • Step D N-(2-(Methoxymethyl)-2-methyl-6-morpholino-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Step F 1-(5-Amino-2-methoxy-4-morpholinophenyl)propan-2-one
  • Step G (R)—N-(3-hydroxy-3-methyl-7-morpholinochroman-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide and (S)—N-(3-hydroxy-3-methyl-7-morpholinochroman-6-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide
  • Peak 1 (37 mg, 2.5%): Purified via Chiral SFC (Thar 350 SFC (Lux Cellulose-1, 30 ⁇ 250 mm, 5 um), 40% MeOH isocratic (0.10% NH 4 OH) in CO2).
  • MS (ESI): m/z 410.2 [M+1] + .
  • reaction mixture was filtered through a plug of silica, washed with a solution of 20% methanol in DCM. The filtrate was then concentrated and purified and resolved via chiral SFC (PIC 100 SFC (Lux Cellulose-3, 21.1*150 mm, 5 um), 25% MeOH isocratic (0.1% NH 4 OH) in CO2) to afford the title compounds as solids. Stereochemistry was arbitrarily assigned based on peak elution.
  • N-(2-(hydroxymethyl)-2-methyl-6-morpholino-2,3-dihydrobenzofuran-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (Example 6) was resolved via chiral SFC (PIC 100 SFC (Lux Cellulose-3, 21.1*150 mm, 5 um), 25% MeOH isocratic (0.1% NH 4 OH) in CO 2 ) to afford the title compounds as solids. Stereochemistry was arbitrarily assigned based on peak elution.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
US16/014,231 2015-12-22 2018-06-21 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS Abandoned US20180298015A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/014,231 US20180298015A1 (en) 2015-12-22 2018-06-21 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
US16/775,650 US10988478B1 (en) 2015-12-22 2020-01-29 Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators
US17/172,766 US20220332718A1 (en) 2015-12-22 2021-02-10 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201562271171P 2015-12-22 2015-12-22
US201662279459P 2016-01-15 2016-01-15
US201662398341P 2016-09-22 2016-09-22
PCT/EP2016/081810 WO2017108723A2 (en) 2015-12-22 2016-12-19 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
US16/014,231 US20180298015A1 (en) 2015-12-22 2018-06-21 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/081810 Continuation WO2017108723A2 (en) 2015-12-22 2016-12-19 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/775,650 Continuation US10988478B1 (en) 2015-12-22 2020-01-29 Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators

Publications (1)

Publication Number Publication Date
US20180298015A1 true US20180298015A1 (en) 2018-10-18

Family

ID=57755272

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/014,231 Abandoned US20180298015A1 (en) 2015-12-22 2018-06-21 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
US16/775,650 Active US10988478B1 (en) 2015-12-22 2020-01-29 Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators
US17/172,766 Abandoned US20220332718A1 (en) 2015-12-22 2021-02-10 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/775,650 Active US10988478B1 (en) 2015-12-22 2020-01-29 Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators
US17/172,766 Abandoned US20220332718A1 (en) 2015-12-22 2021-02-10 PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS

Country Status (7)

Country Link
US (3) US20180298015A1 (zh)
EP (1) EP3394066A2 (zh)
JP (1) JP6895439B2 (zh)
CN (1) CN108473498B (zh)
HK (1) HK1259623A1 (zh)
TW (1) TW201726680A (zh)
WO (1) WO2017108723A2 (zh)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019111218A1 (en) 2017-12-08 2019-06-13 Cadila Healthcare Limited Novel heterocyclic compounds as irak4 inhibitors
US10336762B2 (en) 2017-02-16 2019-07-02 Gilead Sciences, Inc. Pyrrolo[1,2-b]pyridazine derivatives
US10875866B2 (en) 2018-07-13 2020-12-29 Gilead Sciences, Inc. Pyrrolo[1,2-B]pyridazine derivatives
WO2023076556A1 (en) * 2021-10-29 2023-05-04 Kymera Therapeutics, Inc. Irak4 degraders and synthesis thereof
WO2024076614A3 (en) * 2022-10-04 2024-05-16 Children's Hospital Medical Center Multi-cyclic irak and flt3 inhibiting compounds and uses thereof

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA119835C2 (uk) 2016-02-24 2019-08-12 Пфайзер Інк. ПОХІДНІ ПІРАЗОЛО[1,5-а]ПІРАЗИН-4-ІЛУ ЯК JAK-ІНГІБІТОРИ
CN109890829B (zh) * 2016-11-02 2022-07-15 豪夫迈·罗氏有限公司 作为IRAK4调节剂的吡唑并[1,5a]嘧啶衍生物
JP2020524663A (ja) 2017-06-21 2020-08-20 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Irak4調節因子としてのイソインドリノン誘導体
CN110770229A (zh) * 2017-06-21 2020-02-07 豪夫迈·罗氏有限公司 作为irak4调节剂的苯并呋喃化合物
EP3642204A1 (en) 2017-06-21 2020-04-29 H. Hoffnabb-La Roche Ag PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
WO2019034973A1 (en) 2017-08-14 2019-02-21 Pfizer Inc. PYRAZOLO [1,5-A] PYRAZIN-4-YL AND RELATED DERIVATIVES
US11358948B2 (en) 2017-09-22 2022-06-14 Kymera Therapeutics, Inc. CRBN ligands and uses thereof
IL295603B2 (en) 2017-09-22 2024-03-01 Kymera Therapeutics Inc Protein compounds and their uses
IL275649B2 (en) 2017-12-26 2023-12-01 Kymera Therapeutics Inc IRAK joints and used in them
GB201721961D0 (en) 2017-12-27 2018-02-07 Bp Oil Int Methods for preparing fuel additives
GB201721957D0 (en) 2017-12-27 2018-02-07 Bp Oil Int Methods for preparing fuel additives
GB201721960D0 (en) * 2017-12-27 2018-02-07 Bp Oil Int Methods for preparing fuel additives
GB201721964D0 (en) 2017-12-27 2018-02-07 Bp Oil Int Methods for preparing fuel additives
GB201721967D0 (en) 2017-12-27 2018-02-07 Bp Oil Int Methods for preparing fuel additives
WO2019140380A1 (en) 2018-01-12 2019-07-18 Kymera Therapeutics, Inc. Protein degraders and uses thereof
EP3737675A4 (en) 2018-01-12 2022-01-05 Kymera Therapeutics, Inc. CRBN LIGANDS AND THEIR USES
CN108586464A (zh) * 2018-04-12 2018-09-28 苏州康润医药有限公司 一种3-溴吡唑并[1,5-α]嘧啶-6-甲酸的合成方法
CN108911987A (zh) * 2018-05-25 2018-11-30 江苏道博化工有限公司 一种5-硝基-间苯二甲酸的合成工艺
EP3817748A4 (en) 2018-07-06 2022-08-24 Kymera Therapeutics, Inc. TRICYCLIC CRBN LIGANDS AND USES THEREOF
CN113423427A (zh) 2018-11-30 2021-09-21 凯麦拉医疗公司 Irak降解剂和其用途
EP3670501A1 (en) 2018-12-17 2020-06-24 Basf Se Substituted [1,2,4]triazole compounds as fungicides
GB201904375D0 (en) * 2019-03-29 2019-05-15 Galapagos Nv Novel compounds and pharmaceutical compositions thereof for the treatment of inflammatory disorders
WO2021127283A2 (en) 2019-12-17 2021-06-24 Kymera Therapeutics, Inc. Irak degraders and uses thereof
IL293917A (en) 2019-12-17 2022-08-01 Kymera Therapeutics Inc Iraq joints and their uses
TW202210483A (zh) 2020-06-03 2022-03-16 美商凱麥拉醫療公司 Irak降解劑之結晶型
EP4223750A4 (en) * 2020-09-30 2024-06-19 Asahi Kasei Pharma Corporation NITROGEN-CONTAINING BICYCLIC COMPOUND WITH PYRIMIDINE
WO2022070288A1 (ja) * 2020-09-30 2022-04-07 旭化成ファーマ株式会社 含窒素二環化合物
AU2022421224A1 (en) * 2021-12-22 2024-06-20 The Regents Of The University Of California Covalently binding inhibitors of g12s, g12d and/or g12e mutants of k-ras gtpase
TW202409023A (zh) 2022-07-14 2024-03-01 美商富曼西公司 除草苯并𠯤
CN117209489A (zh) * 2023-05-30 2023-12-12 杭州邦顺制药有限公司 Irak激酶抑制剂

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0329214D0 (en) * 2003-12-17 2004-01-21 Glaxo Group Ltd Novel compounds
EP2219646A4 (en) * 2007-12-21 2010-12-22 Univ Rochester METHOD FOR MODIFYING THE LIFETIME OF EUKARYOTIC ORGANISMS
JP5985473B2 (ja) 2010-07-13 2016-09-06 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft IRAK4モジュレーターとしてのピラゾロ[1,5a]ピリミジン及びチエノ[3,2b]ピリミジン誘導体
CA2952188A1 (en) * 2014-06-20 2015-12-23 Aurigene Discovery Technologies Limited Substituted indazole compounds as irak4 inhibitors
JP6734860B2 (ja) * 2014-11-06 2020-08-05 リソソーマル・セラピューティクス・インコーポレイテッドLysosomal Therapeutics Inc. 置換ピラゾロ[1,5−a]ピリミジンおよび医療疾患の治療におけるその使用
CN104667646B (zh) * 2015-01-23 2017-02-22 北京博源恒升高科技有限公司 一种废气除尘方法及其装置
BR112018000624A2 (pt) * 2015-07-15 2018-09-18 Aurigene Discovery Technologies Limited compostos de indazol e azaindazol como inibidores de irak-4

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10336762B2 (en) 2017-02-16 2019-07-02 Gilead Sciences, Inc. Pyrrolo[1,2-b]pyridazine derivatives
WO2019111218A1 (en) 2017-12-08 2019-06-13 Cadila Healthcare Limited Novel heterocyclic compounds as irak4 inhibitors
US10875866B2 (en) 2018-07-13 2020-12-29 Gilead Sciences, Inc. Pyrrolo[1,2-B]pyridazine derivatives
US11535622B2 (en) 2018-07-13 2022-12-27 Gilead Sciences, Inc. Pyrrolo[1,2-b] pyridazine derivatives
WO2023076556A1 (en) * 2021-10-29 2023-05-04 Kymera Therapeutics, Inc. Irak4 degraders and synthesis thereof
WO2024076614A3 (en) * 2022-10-04 2024-05-16 Children's Hospital Medical Center Multi-cyclic irak and flt3 inhibiting compounds and uses thereof

Also Published As

Publication number Publication date
CN108473498B (zh) 2021-11-02
EP3394066A2 (en) 2018-10-31
US10988478B1 (en) 2021-04-27
CN108473498A (zh) 2018-08-31
JP2019503357A (ja) 2019-02-07
JP6895439B2 (ja) 2021-06-30
HK1259623A1 (zh) 2019-12-06
US20220332718A1 (en) 2022-10-20
WO2017108723A2 (en) 2017-06-29
TW201726680A (zh) 2017-08-01
WO2017108723A3 (en) 2017-07-27

Similar Documents

Publication Publication Date Title
US10988478B1 (en) Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators
US10899772B2 (en) IRAK4 modulators
US20220402915A1 (en) Bcl-2 inhibitors
EP3145929B1 (en) 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds which are jak inhibitors
US11034698B2 (en) Pyrazolo[ 1,5a]pyrimidine derivatives as IRAK4 modulators
EP3642199B1 (en) Benzofurans as irak4 modulators
JP2018530557A (ja) 置換ピラゾロ[3,4−b]ピリジン−6−カルボン酸およびこの使用
US11434250B2 (en) Pyrazolo[1,5a]pyrimidine derivatives as IRAK4 modulators
CN116829151A (zh) 氮杂喹唑啉泛KRas抑制剂
WO2023131122A1 (zh) 稠环取代的六元杂环化合物及其制法和用途

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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