USRE47929E1 - Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses - Google Patents

Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses Download PDF

Info

Publication number
USRE47929E1
USRE47929E1 US16/201,653 US201316201653A USRE47929E US RE47929 E1 USRE47929 E1 US RE47929E1 US 201316201653 A US201316201653 A US 201316201653A US RE47929 E USRE47929 E US RE47929E
Authority
US
United States
Prior art keywords
patient
mmol
acceptable salt
administering
treating
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.)
Active
Application number
US16/201,653
Other languages
English (en)
Inventor
Ryan M. Moslin
David S. Weinstein
Stephen T. Wrobleski
John S. Tokarski
Shuqun Lin
Steven H. Spergel
Yanlei Zhang
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=49876960&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE47929(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US16/201,653 priority Critical patent/USRE47929E1/en
Application granted granted Critical
Publication of USRE47929E1 publication Critical patent/USRE47929E1/en
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIN, SHUQUN, SPERGEL, STEVEN H., ZHANG, YANLEI
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WROBLESKI, STEPHEN T., TOKARSKI, JOHN S., WEINSTEIN, DAVID S., MOSLIN, Ryan M.
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • C07B59/002Heterocyclic compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D237/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • This invention relates to compounds useful in the modulation of IL-12, IL-23 and/or IFN ⁇ by acting on Tyk-2 to cause signal transduction inhibition.
  • amide-substituted heterocyclic compounds Provided herein are amide-substituted heterocyclic compounds, compositions comprising such compounds, and methods of their use.
  • the invention further pertains to pharmaceutical compositions containing at least one compound according to the invention that are useful for the treatment of conditions related to the modulation of IL-12, IL-23 and/or IFN ⁇ in a mammal.
  • the heterodimeric cytokines interleukin (IL)-12 and IL-23 which share a common p40 subunit, are produced by activated antigen-presenting cells and are critical in the differentiation and proliferation of Th1 and Th17 cells, two effector T cell lineages which play key roles in autoimmunity.
  • IL-23 is composed of the p40 subunit along with a unique p19 subunit.
  • IL-23, acting through a heterodimeric receptor composed of IL-23R and IL-12R ⁇ 1 is essential for the survival and expansion of Th17 cells which produce pro-inflammatory cytokines such as IL-17A, IL-17F, IL-6 and TNF- ⁇ (McGeachy, M. J.
  • IL-12 in addition to the p40 subunit in common with IL-23, contains a p35 subunit and acts through a heterodimeric receptor composed of IL-12R ⁇ 1 and IL-12R ⁇ 2.
  • IL-12 is essential for Th1 cell development and secretion of IFN ⁇ , a cytokine which plays a critical role in immunity by stimulating MHC expression, class switching of B cells to IgG subclasses, and the activation of macrophages (Gracie, J. A. et al., “Interleukin-12 induces interferon-gamma-dependent switching of IgG alloantibody subclass”, Eur. J. Immunol., 26:1217-1221 (1996); Schroder, K. et al., “Interferon-gamma: an overview of signals, mechanisms and functions”, J. Leukoc. Biol., 75(2):163-189 (2004)).
  • mice deficient in either p40, p19, or IL-23R are protected from disease in models of multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, lupus and psoriasis, among others (Kyttaris, V. C. et al., “Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice”, J. Immunol., 184:4605-4609 (2010); Hong, K.
  • IL-12 independently of IFN-gamma, plays a crucial role in the pathogenesis of a murine psoriasis like skin disorder”, J. Immunol., 162:7480-7491 (1999); Hue, S. et al., “Interleukin-23 drives innate and T cell-mediated intestinal inflammation”, J. Exp. Med., 203:2473-2483 (2006); Cua, D. J. et al., “Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain”, Nature, 421:744-748 (2003); Murphy, C. A. et al., “Divergent pro- and anti-inflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation”, J. Exp. Med., 198:1951-1957 (2003)).
  • Th17 cells have been identified in active lesions in the brain from MS patients and in the gut mucosa of patients with active Crohn's disease (Lee, E. et al., “Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris”, J. Exp. Med., 199:125-130 (2004); Tzartos, J. S. et al., “Interleukin-17 production in central nervous system infiltrating T cells and glial cells is associated with active disease in multiple sclerosis”, Am. J. Pathol., 172:146-155 (2008)).
  • mRNA levels of p19, p40, and p35 in active SLE patients were also shown to be significantly higher compared with those in inactive SLE patients (Huang, X. et al., “Dysregulated expression of interleukin-23 and interleukin-12 subunits in systemic lupus erythematosus patients”, Mod. Rheumatol., 17:220-223 (2007)), and T cells from lupus patients have a predominant Th1 phenotype (Tucci, M. et al., “Overexpression of interleukin-12 and T helper 1 predominance in lupus nephritis”, Clin. Exp. Immunol., 154:247-254 (2008)).
  • anti-p40 treatment which inhibits both IL-12 and IL-23, as well as IL-23-specific anti-p19 therapies have been shown to be efficacious in the treatment of autoimmunity in diseases including psoriasis, Crohn's Disease and psoriatic arthritis (Leonardi, C. L. et al., “PHOENIX 1 study investigators. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomized, double-blind, placebo-controlled trial (PHOENIX 1)”, Lancet, 371:1665-1674 (2008); Sandborn, W. J.
  • Type I group of interferons which include the IFN ⁇ members as well as IFN ⁇ , IFN ⁇ , IFN ⁇ and IFN ⁇ , act through a heterodimer IFN ⁇ / ⁇ receptor (IFNAR).
  • Type I IFNs have multiple effects in both the innate and adaptive immune systems including activation of both the cellular and humoral immune responses as well as enhancing the expression and release of autoantigens (Hall, J. C. et al., “Type I interferons: crucial participants in disease amplification in autoimmunity”, Nat. Rev. Rheumatol., 6:40-49 (2010)).
  • IFN ⁇ interferon
  • IFN ⁇ signature type I IFN-regulated genes
  • IFN ⁇ A direct role for IFN ⁇ in the pathobiology of lupus is evidenced by the observation that the administration of IFN ⁇ to patients with malignant or viral diseases can induce a lupus-like syndrome. Moreover, the deletion of the IFNAR in lupus-prone mice provides high protection from autoimmunity, disease severity and mortality (Santiago-Raber, M. L. et al., “Type-I interferon receptor deficiency reduces lupus-like disease in NZB mice”, J. Exp.
  • Tyrosine kinase 2 (Tyk2) is a member of the Janus kinase (JAK) family of nonreceptor tyrosine kinases and has been shown to be critical in regulating the signal transduction cascade downstream of receptors for IL-12, IL-23 and type I interferons in both mice (Ishizaki, M. et al., “Involvement of Tyrosine Kinase-2 in Both the IL-12/Th1 and IL-23/Th17 Axes In vivo”, J. Immunol., 187:181-189 (2011); Prchal-Murphy, M.
  • J. Immunol. 187:181-189 (2011)
  • Prchal-Murphy M.
  • Tyk2 mediates the receptor-induced phosphorylation of members of the STAT family of transcription factors, an essential signal that leads to the dimerization of STAT proteins and the transcription of STAT-dependent pro-inflammatory genes.
  • Tyk2-deficient mice are resistant to experimental models of colitis, psoriasis and multiple sclerosis, demonstrating the importance of Tyk2-mediated signaling in autoimmunity and related disorders (Ishizaki, M. et al., “Involvement of Tyrosine Kinase-2 in Both the IL-12/Th1 and IL-23/Th17 Axes In vivo”, J. Immunol., 187:181-189 (2011); Oyamada, A. et al., “Tyrosine kinase 2 plays critical roles in the pathogenic CD4 T cell responses for the development of experimental autoimmune encephalomyelitis”, J. Immunol., 183:7539-7546 (2009)).
  • Tyk2 In humans, individuals expressing an inactive variant of Tyk2 are protected from multiple sclerosis and possibly other autoimmune disorders (Couturier, N. et al., “Tyrosine kinase 2 variant influences T lymphocyte polarization and multiple sclerosis susceptibility”, Brain, 134:693-703 (2011)). Genome-wide association studies have shown other variants of Tyk2 to be associated with autoimmune disorders such as Crohn's Disease, psoriasis, systemic lupus erythematosus, and rheumatoid arthritis, further demonstrating the importance of Tyk2 in autoimmunity (Ellinghaus, D.
  • new compounds capable of modulating cytokines and/or interferons such as IL-12, IL-23 and/or IFN ⁇ , and methods of using these compounds may provide substantial therapeutic benefits to a wide variety of patients in need thereof.
  • the invention is directed to compounds of Formula I, infra, that which are useful as modulators of IL-12, IL-23 and/or IFN ⁇ by inhibiting Tyk2-mediated signal transduction.
  • the present invention also provides processes and intermediates for making the compounds of the present invention.
  • the present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention.
  • the present invention also provides a method for the modulation of IL-12, IL-23 and/or IFN ⁇ by inhibiting Tyk-2-mediated signal transduction comprising administering to a host in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention.
  • the present invention also provides a method for treating proliferative, metabolic, allergic, autoimmune and inflammatory diseases, comprising administering to a host in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention.
  • a preferred embodiment is a method for treating inflammatory and autoimmune diseases or diseases.
  • an inflammatory and autoimmune disease or disorder includes any disease having an inflammatory or autoimmune component.
  • An alternate preferred embodiment is a method for treating metabolic diseases, including type 2 diabetes and atherosclerosis.
  • the present invention also provides the use of the compounds of the present invention for the manufacture of a medicament for the treatment of cancers.
  • the present invention also provides the compounds of the present invention for use in therapy.
  • Y is N or CR 6 CR 6 ;
  • R 1 is H, C 1-3 alkyl or C 3-6 cycloalkyl, each optionally substituted by 0-7 R 1a ;
  • R 1a at each occurrence is independently hydrogen, deuterium, F, Cl, Br or CN;
  • R 2 is C 1-6 alkyl, —(CH 2 ) r -3-14 membered carbocycle substituted with 0-1 R 2a or a 5-14 membered heterocycle containing 1-4 heteroatoms selected from N, O, and S, each group substituted with 0-4 R 2a (for the sake of clarity, R 2 is intended to include substituted methyl groups such as —C(O)R 2a );
  • R 2a at each occurrence is independently hydrogen, ⁇ O, halo, OCF 3 , CN, NO 2 , —(CH 2 ) r OR b , —(CH 2 ) r SR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r C(O)OR b , —(CH 2 ) r OC(O)R b , CH 2 ) r NR 11 R 11 , —(CH 2 ) r C(O)NR 11 R 11 , —(CH 2 ) r NR b C(O)R c , —(CH 2 ) r NR b C(O)OR c , —NR b C(O)NR 11 R 11 , —S(O) p NR 11 R 11 , —NR b S(O) p R c , —S(O) p R c , C 1-6 al
  • R 3 is C 3-10 cycloalkyl, C 6-10 aryl or a 5-10 membered heterocycle containing 1-4 heteroatoms selected from N, O, and S, each group substituted with 0-4 R 3a ;
  • R 3a at each occurrence is independently hydrogen, ⁇ O, halo, OCF 3 , CF 3 , CHF 2 , CN, NO 2 , —(CH 2 ) r OR b , —(CH 2 ) r SR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r C(O)OR b , —(CH 2 ) r OC(O)R b , —(CH 2 ) r NR 11 R 11 , —(CH 2 ) r C(O)NR 11 R 11 , —(CH 2 ) r NR b C(O)R c , —(CH 2 ) r NR b C(O)OR c , —NR b C(O)NR 11 R 11 , —S(O) p NR 11 R 11 , —NR b S(O) p R c , —S(
  • R 3a together with the atoms to which they are attached, combine to form a fused ring wherein said ring is selected from phenyl and a heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p , each fused ring substituted with 0-3 R a1 ;
  • R 4 and R 5 are independently hydrogen, C 1-4 alkyl substituted with 0-1 R f , (CH 2 ) r- phenyl substituted with 0-3 R d or a —(CH 2 )-5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p ;
  • R 6 is hydrogen, halo, C 1-4 alkyl, C 1-4 haloalkyl, OC 1-4 haloalkyl, OC 1-4 alkyl, CN, NO 2 or OH;
  • R 11 at each occurrence is independently hydrogen, C 1-4 alkyl substituted with 0-3 R f , CF 3 , C 3-10 cycloalkyl substituted with 0-1 R f , (CH) r -phenyl substituted with 0-3 R d or —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-3 R d ;
  • R a and R a1 at each occurrence are independently hydrogen, F, Cl, Br, OCF 3 , CF 3 , CHF 2 , CN, NO 2 , —(CH 2 ) r OR b , —(CH 2 ) r SR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r C(O)OR 6 , —(CH 2 ) r OC(O)R b , —(CH 2 ) r NR 11 R 11 , —(CH 2 ) r C(O)NR 11 R 11 , —(CH 2 ) r NR b C(O)R c , —(CH 2 ) r NR b C(O)OR c , —NR b C(O)NR 11 R 11 , —S(O) p NR“R”, —NR b S(O) p R c , —S(
  • R b is hydrogen, C 1-6 alkyl substituted with 0-3 R d , C 1-6 haloalkyl, C 3-6 cycloalkyl substituted with 0-2 R d , or —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-3 R f or (CH 2 ) r -phenyl substituted with 0-3 R d ;
  • R c is C 1-6 alkyl substituted with 0-3 R f , (CH 2 ) r —C 3-6 cycloalkyl substituted with 0-3 R f or (CH 2 ) r -phenyl substituted with 0-3 R f ;
  • R d at each occurrence is independently hydrogen, F, Cl, Br, OCF 3 , CF 3 , CN, NO 2 , —OR e , —(CH 2 ) r C(O)R c , —NR e R e , —NR e C(O)OR c , C 1-6 alkyl or (CH 2 ) r -phenyl substituted with 0-3 R f ;
  • R e at each occurrence is independently selected from hydrogen, C 1-6 alkyl, C 3-6 cycloalkyl and (CH 2 ) r -phenyl substituted with 0-3 R f ;
  • R f independently at each occurrence is hydrogen, halo, CN, NH 2 , OH, C 3-6 cycloalkyl, CF 3 , O(C 1-6 alkyl) or a —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p ;
  • p 0, 1, or 2;
  • r 0, 1, 2, 3, or 4.
  • R 2 is —C(O)R 2a ; or C 1-6 alkyl, C 3-6 cycloalkyl, phenyl, pyrazolyl, thiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, quinolinyl or pyrrolopyridinyl, each group substituted by 0-4 groups selected from R 2a .
  • R 2 is —C(O)R 2a ; or C 1-6 alkyl, C 3-6 cycloalkyl, or phenyl, each group substituted by 0-4 groups selected from R 2a .
  • R 2 is pyrazolyl, thiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, quinolinyl or pyrrolopyridinyl, each group substituted by 0-4 groups selected from R 2a .
  • R 1 is H or C 1-3 alkyl substituted by 0-7 R 1a ;
  • R 1a at each occurrence is independently hydrogen, deuterium or halogen (preferably H, D or F);
  • R 2 is C 1-6 alkyl, C 3-6 cycloalkyl, phenyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, quinolinyl or pyrrolopyridinyl, each group substituted by 0-4 groups selected from R 2a ;
  • R 2a at each occurrence is independently hydrogen, ⁇ O, halo, CN, —(CH 2 ) r OR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r C(O)NR 11 R 11 , —S(O) p NR 11 R 11 , —C 1-6 alkyl substituted with 0-3 R a , C 1-6 haloalkyl, —(CH 2 ) r -3-14 membered carbocycle substituted with 0-1 R a or a —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-2 R a ;
  • R 3 is C 3-10 cycloalkyl, a C 6-10 aryl, or a 5-10 membered heterocycle containing 1-4 heteroatoms selected from N, O, and S, each group substituted with 0-4 R 3a ;
  • R 3a at each occurrence is independently hydrogen, halo, OCF 3 , CF 3 , CHF 2 , CN, —(CH 2 ) r OR b , —(CH 2 ) r SR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r NR 11 R 11 , —(CH 2 ) r C(O)NR 11 R 11 , —(CH 2 ) r NR b C(O)R c , —S(O) p NR 11 R 11 , —NR b S(O) p R c , —S(O) p R c , C 1-6 alkyl substituted with 0-3 R a , C 1-6 haloalkyl, a —(CH 2 ) r -3-14 membered carbocycle substituted with 0-3 R a or a —(CH 2 ) r -5-10 membere
  • R 3a together with the atoms to which they are attached, combine to form a fused ring wherein that ring is selected from phenyl and a 5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, S or O, each fused ring substituted by 0-3 R a1 ;
  • R 11 at each occurrence is independently hydrogen, C 1-4 alkyl substituted with 0-3 R f or C 3-10 cycloalkyl substituted with 0-1 R f ;
  • R a and R a1 at each occurrence are independently hydrogen, ⁇ O, F, —(CH 2 ) r OR b or C 1-6 alkyl substituted with 0-3 R f ;
  • R b is hydrogen, C 1-6 alkyl substituted with 0-3 R d , C 1-6 haloalkyl, C 3-6 cycloalkyl substituted with 0-2 R d , or —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-3 R f or (CH 2 ) r -phenyl substituted with 0-3 R d ;
  • R c is C 1-6 alkyl substituted with 0-3 R f ;
  • R d at each occurrence is independently hydrogen, halo (preferably F), or —OH;
  • R f at each occurrence is independently hydrogen, halo, CN, OH or O(C 1-6 alkyl);
  • p 0, 1 or 2;
  • r 0, 1 or 2.
  • R 1 is H or C 1-3 alkyl substituted by 0-7 R 1a ;
  • R 1a at each occurrence is independently hydrogen, deuterium or halogen
  • R 2 is —C(O)R 2a ; or C 1-6 alkyl, C 3-6 cycloalkyl, phenyl, pyrazolyl, thiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, quinolinyl or pyrrolopyridinyl, each group substituted by 0-4 groups selected from R 2a ;
  • R 2a at each occurrence is independently hydrogen, ⁇ O, halo, CN, —(CH 2 ) r OR b , —(CH 2 ) r C(O)R b , —NR b C(O)R c , —C(O)OR b , —(CH 2 ) r C(O)NR 11 R 11 , —S(O) p NR 11 R 11 , —C 1-6 alkyl substituted with 0-3 R a , C 1-6 haloalkyl, —(CH 2 ) r -3-14 membered carbocycle substituted with 0-1 R a or a —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-2 R a ;
  • R 3 is C 3-10 cycloalkyl, a C 6-10 aryl, or a 5-10 membered heterocycle containing 1-4 heteroatoms selected from N, O, and S, each group substituted with 0-4 R 3a ;
  • R 3a at each occurrence is independently hydrogen, halo, OCF 3 , CF 3 , CHF 2 , CN, —(CH 2 ) r OR b , —(CH 2 ) r SR b , —(CH 2 ) r C(O)R b , —(CH 2 ) r NR 11 R 11 , —(CH 2 ) r C(O)NR 11 R 11 , —(CH 2 ) r NR b C(O)R c , —S(O) p NR 11 R 11 , —NR b S (O) p R c , —S(O) p R c , C 1-6 alkyl substituted with 0-3 R a , C 1-6 haloalkyl, a —(CH 2 ) r -3-14 membered carbocycle substituted with 0-3 R a or a —(CH 2 ) r -5-10 membere
  • R 3a together with the atoms to which they are attached, combine to form a fused ring wherein that ring is selected from phenyl and a 5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, S or O, each fused ring substituted, as valence allows, by 0-3 R a ;
  • R 11 at each occurrence is independently hydrogen, C 1-4 alkyl substituted with 0-3 R f or C 3-6 cycloalkyl substituted with 0-1 R f ;
  • R a at each occurrence is hydrogen, ⁇ O, F, —(CH 2 ) r OR b or C 1-6 alkyl substituted with 0-3 R f ;
  • R b is hydrogen, C 1-6 alkyl substituted with 0-3 R d , C 1-6 haloalkyl, C 3-6 cycloalkyl substituted with 0-2 R d , or —(CH 2 ) r -5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, O, and S(O) p substituted with 0-3 R f or (CH 2 ) r -phenyl substituted with 0-3 R d ;
  • R c is C 1-6 alkyl or C 3-6 cycloalkyl, each group substituted with 0-3 R f ;
  • R d at each occurrence is independently hydrogen, F, Cl, Br or —OH;
  • R f at each occurrence is independently hydrogen, halo, CN, OH or O(C 1-6 alkyl);
  • p 0, 1 or 2;
  • r 0, 1 or 2.
  • a compound of formula I or a stereoisomer or pharmaceutically-acceptable salt thereof, wherein R 2 is pyrazolyl, thiazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl or quinolinyl, each group substituted with 0-3 R 2a (especially preferred embodiments are those wherein R 2a is halo, CN or phenyl).
  • a compound of formula I or a stereoisomer or pharmaceutically-acceptable salt thereof, wherein R 2 is —C(O)R 2a ; or C 1-6 alkyl, C 3-6 cycloalkyl or phenyl, each group substituted with 0-3 R 2a .
  • R 2 is selected from:
  • a compound of formula (I), or a stereoisomer or pharmaceutically-acceptable salt thereof wherein R 3 is phenyl, cyclopentyl, cyclohexyl, furanyl, or pyranyl, each substituted with 0-4 R 3a (preferably, R 3 is phenyl substituted with 0-3 R 3a ).
  • R 3a at each occurrence independently is hydrogen, Ph, CN, NH 2 , OCF 3 , OR b , halo, cycloalkyl, C(O)NR 11 R 11 , S(O) 2 NR 11 R 11 , C(O)R b , SO p R c , NR b SO p R c , NR b C(O)R c , haloalkyl, CN, 5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, S or O substituted with 0-3 R a and C 1-6 alkyl substituted with 0-3 R a ; or
  • R 3a and a second R 3a together with the atoms to which they are attached, combine to form a fused 5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, S or O or phenyl;
  • R 11 at each occurrence independently is hydrogen, C 3-6 cycloalkyl substituted with 0-3 R f , or C 1-4 alkyl substituted with 0-1 R f ;
  • R a independently at each occurrence is C 1-6 alkyl substituted with 0-3 R f , halo (F) or OR b ;
  • R b independently at each occurrence is hydrogen, 5-7 membered heterocycle comprising carbon atoms and 1-4 heteroatoms selected from N, S or O substituted with 0-3 R f , or C 1-6 alkyl substituted with 0-3 R d ;
  • R d independently at each occurrence is F, Cl, Br or OH;
  • R c independently at each occurrence is C 1-6 alkyl or C 3-6 cycloalkyl, each group substituted with 0-3 R f substituted with 0-3 R f ;
  • R f independently at each occurrence is hydrogen, halo or OH
  • R 3aa is S(O) p R c , OR b , chloro, F, CN, NH 2 , C(O)NR 11 R 11 , NR b SO p R c , NR b C(O)R c , C 1-6 alkyl substituted with 0-3 R a or a 5- to 6-membered heteroaryl containing 1-3 heteroatoms selected from N, O, and S substituted with 0-3 R 3a ; (especially, R 3aa is S(O) 2 Me or OMe);
  • R 3ab , R 3ac , or R 3ad are independently hydrogen, Cl, F, Br, CN, OR b , C 1-6 alkyl substituted 0-3 R a ; C(O)NR 11 R 11 , C(O)R b , S(O) p R c , or a 4-7 membered heterocycle containing 1-3 heteroatoms selected from N, O, and S substituted with 0-3 R a ; (especially R 3ab , R 3ac , or R 3ad are independently, hydrogen or 5-6 membered heterocycle containing 1-3 heteroatoms selected from N, O, and S substituted with 0-2 R a ;
  • R 11 at each occurrence independently is hydrogen, cyclopropyl substituted with 0-3 R f or C 1-4 alkyl substituted with 0-3 R f ;
  • R a at each occurrence independently is C 1-6 alkyl substituted with 0-3 R f , OR b or halo;
  • R b at each occurrence independently is hydrogen, C 1-6 alkyl substituted with 0-2 R d or a 5- to 7-membered heterocycle containing 1-3 heteroatoms selected from N, O and S;
  • R c at each occurrence independently is C 1-6 alkyl substituted with 0-3 R f ;
  • R d at each occurrence independently is F or OH
  • R f at each occurrence independently is halo or OH
  • p 0-2.
  • R 1 is CH 3 or CD 3 ;
  • R 2 is —C(O)C 3-6 cycloalkyl substituted by 0-2 groups selected from C 1-3 alkyl and halo;
  • R 3aa is —O(C 1-3 alkyl)
  • R 3ab is a triazolyl or tetrazolyl group optionally substituted with C 1-6 alkyl substituted by 0-4 groups selected from F, Cl, or Br
  • R 3ac and R 3ad are both hydrogen.
  • R 3aa is S(O) p R c or C(O)NR 11 R 11 (more preferably R 3aa is SO 2 CH 3 ).
  • R 3aa is S(O) p R c or C(O)NR 11 R 11 (more preferably R 3aa is SO 2 CH 3 or C(O)NH 2 ).
  • R 3aa is OR b . More preferably R 3aa is OH, OMe, OCF 3 , OCHF 2 , OCH 2 F or OEt. Even more preferably, R 3aa is OMe.
  • a compound of formula I or a stereoisomer or pharmaceutically-acceptable salt thereof, wherein R 1 is H, CH 3 , C 2 H 5 , cyclopropyl, CD 3 , or CD 2 CD 3 (preferably CH 3 or CD 3 ).
  • composition comprising one or more compounds of formula I and a pharmaceutically acceptable carrier or diluent.
  • the present invention is also directed to pharmaceutical compositions useful in treating diseases associated with the modulation of IL-12, IL-23 and/or IFN ⁇ by acting on Tyk-2 to cause signal transduction inhibition, comprising compounds of formula I, or pharmaceutically-acceptable salts thereof, and pharmaceutically-acceptable carriers or diluents.
  • the invention further relates to methods of treating diseases associated with the modulation of IL-12, IL-23, and/or IFN ⁇ , comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound according to formula I.
  • the present invention also provides processes and intermediates for making the compounds of the present invention.
  • the present invention also provides a method for treating proliferative, metabolic, allergic, autoimmune and inflammatory diseases (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these diseases), comprising administering to a host in need of such treatment a therapeutically effective amount of at least one of the compounds of the present invention.
  • the present invention also provides a method of treating an inflammatory or autoimmune disease (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these diseases) comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of Formula I.
  • the present invention also provides a method for treating a disease (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these diseases), comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of Formula I, wherein the disease is rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus (SLE), lupus nephritis, cutaneous lupus, inflammatory bowel disease, psoriasis, Crohn's Disease, psoriatic arthritis, Sjögren's syndrome, systemic scleroderma, ulcerative colitis, Graves' disease, discoid lupus erythematosus, adult onset Stills, systemic onset juvenile idiopathic arthritis, gout, gouty arthritis, type 1 diabetes, insulin dependent diabetes mellitus, sepsis, septic shock, Shigellosis, pancreatitis (a
  • the present invention also provides a method of treating an inflammatory or autoimmune disease (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of said diseases), comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of Formula I, wherein the disease is selected from systemic lupus erythematosus (SLE), lupus nephritis, cutaneous lupus, Crohn's Disease, ulcerative colitis, type 1 diabetes, psoriasis, rheumatoid arthritis, systemic onset juvenile idiopathic arthritis, ankylosing spondylitis, and multiple sclerosis.
  • SLE systemic lupus erythematosus
  • lupus nephritis cutaneous lupus
  • Crohn's Disease ulcerative colitis
  • type 1 diabetes psoriasis
  • rheumatoid arthritis systemic onset juvenile idiopathic arthritis
  • the present invention also provides a method for treating a rheumatoid arthritis (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of rheumatoid arthritis, comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of Formula I.
  • the present invention also provides a method of treating a condition (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these conditions) comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of Formula I, wherein the condition is selected from acute myelogenous leukemia, chronic myelogenous leukemia, metastatic melanoma, Kaposi's sarcoma, multiple myeloma, solid tumors, ocular neovasculization, and infantile haemangiomas, B cell lymphoma, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, multiple vasculitides, idiopathic thrombocytopenic purpura (ITP), myasthenia gravis, allergic rhinitis, multiple sclerosis (MS), transplant rejection, Type I diabetes, membranous nephritis, inflammatory a,
  • the present invention also provides a method of treating a IL-12, IL-23, and/or IFN ⁇ mediated disease (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these diseases), comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of formula I.
  • the present invention also provides a method of treating a IL-12, IL-23 and/or IFN ⁇ mediated disease (or use of the compounds of the present invention for the manufacture of a medicament for the treatment of these diseases), comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of formula I, wherein the IL-12, IL-23 and/or IFN ⁇ mediated disease is a disease modulated by IL-12, IL-23 and/or IFN ⁇ .
  • the present invention also provides a method of treating diseases, comprising administering to a patient in need of such treatment a therapeutically-effective amount of a compound of formula I in combination with other therapeutic agents.
  • the present invention also provides the compounds of the present invention for use in therapy.
  • compounds of formula I are selected from exemplified compounds or combinations of exemplified compounds or other embodiments herein.
  • Compounds of this invention may have one or more asymmetric centers. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms of compounds of the present invention are included in the present invention. Many geometric isomers of olefins, C ⁇ N double bonds, and the like can also be present in the compounds, and all such stable isomers are contemplated in the present invention. Cis- and trans-geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. The present compounds can be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis from optically active starting materials. All chiral, (enantiomeric and diastereomeric) and racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomer form is specifically indicated.
  • any variable e.g., R 3
  • its definition at each occurrence is independent of its definition at every other occurrence.
  • R 3 at each occurrence is selected independently from the definition of R 3 .
  • combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • nitrogen atoms e.g., amines
  • these can be converted to N-oxides by treatment with an oxidizing agent (e.g., MCPBA and/or hydrogen peroxides) to afford other compounds of this invention.
  • an oxidizing agent e.g., MCPBA and/or hydrogen peroxides
  • a dash “-” that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, —CONH 2 is attached through the carbon atom.
  • optionally substituted in reference to a particular moiety of the compound of Formula I (e.g., an optionally substituted heteroaryl group) refers to a moiety having 0, 1, 2, or more substituents.
  • optionally substituted alkyl encompasses both “alkyl” and “substituted alkyl” as defined below. It will be understood by those skilled in the art, with respect to any group containing one or more substituents, that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical, synthetically non-feasible and/or inherently unstable.
  • alkyl or “alkylene” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.
  • C 1-10 alkyl (or alkylene) is intended to include C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , and C 10 alkyl groups.
  • C 1 -C 6 alkyl denotes alkyl having 1 to 6 carbon atoms.
  • Alkyl groups can be unsubstituted or substituted so that one or more of its hydrogens are replaced by another chemical group.
  • Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t-butyl), pentyl (e.g., n-pentyl, isopentyl, neopentyl), and the like.
  • alkenyl or “alkenylene” is intended to include hydrocarbon chains of either straight or branched configuration and having one or more double carbon-carbon bonds that may occur in any stable point along the chain.
  • C 2-6 alkenyl (or alkenylene), is intended to include C 2 , C 3 , C 4 , C 5 , and C 6 alkenyl groups.
  • alkenyl examples include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-methyl-2-propenyl, 4-methyl-3-pentenyl, and the like.
  • Alkynyl or “alkynylene” is intended to include hydrocarbon chains of either straight or branched configuration and having one or more triple carbon-carbon bonds that may occur in any stable point along the chain.
  • C 2-6 alkynyl (or alkynylene), is intended to include C 2 , C 3 , C 4 , C 5 , and C 6 alkynyl groups; such as ethynyl, propynyl, butynyl, pentynyl, hexynyl and the like.
  • alkyl refers to a substituted alkyl group as defined above where at least one of the substituents is an aryl, such as benzyl.
  • aryl(C 0-4 )alkyl includes a substituted lower alkyl having at least one aryl substituent and also includes an aryl directly bonded to another group, i.e., aryl(C 0 )alkyl.
  • heteroarylalkyl refers to a substituted alkyl group as defined above where at least one of the substituents is a heteroaryl.
  • substituted alkenyl, alkynyl, alkylene, alkenylene, or alkynylene group these groups are substituted with one to three substituents as defined above for substituted alkyl groups.
  • alkoxy refers to an oxygen atom substituted by alkyl or substituted alkyl, as defined herein.
  • alkoxy includes the group —O—C 1-6 alkyl such as methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyloxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, 3-methylpentoxy, and the like.
  • “Lower alkoxy” refers to alkoxy groups having one to four carbons.
  • substituted means that any one or more hydrogens on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded.
  • a substituent is oxo, or keto, (i.e., ⁇ O) then 2 hydrogens on the atom are replaced.
  • Keto substituents are not present on aromatic moieties.
  • substituents are named into the core structure. For example, it is to be understood that when (cycloalkyl)alkyl is listed as a possible substituent, the point of attachment of this substituent to the core structure is in the alkyl portion.
  • Ring double bonds as used herein, are double bonds that are formed between two adjacent ring atoms (e.g., C ⁇ C, C ⁇ N, or N ⁇ N).
  • a stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture to a useful degree of purity, and subsequent formulation into an efficacious therapeutic agent. It is preferred that the presently recited compounds do not contain a N-halo, S(O) 2 H, or S(O)H group.
  • cycloalkyl refers to cyclized alkyl groups, including mono-, bi- or poly-cyclic ring systems.
  • C 3-7 cycloalkyl is intended to include C 3 , C 4 , C 5 , C 6 , and C 7 cycloalkyl groups.
  • Example cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, and the like.
  • “carbocycle” or “carbocyclic residue” is intended to mean any stable 3-, 4-, 5-, 6-, or 7-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, or 13-membered bicyclic or tricyclic ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic.
  • carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, anthracenyl, and tetrahydronaphthyl (tetralin).
  • bridged rings are also included in the definition of carbocycle (e.g., [2.2.2]bicyclooctane).
  • carbocycles e.g., [2.2.2]bicyclooctane
  • Preferred carbocycles are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and phenyl.
  • carbocycle When the term “carbocycle” is used, it is intended to include “aryl”.
  • a bridged ring occurs when one or more carbon atoms link two non-adjacent carbon atoms.
  • Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a bicyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.
  • aryl refers to monocyclic or bicyclic aromatic hydrocarbon groups having 6 to 12 carbon atoms in the ring portion, such as phenyl, and naphthyl groups, each of which may be substituted.
  • cycloalkyl includes cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclooctyl, etc., as well as the following ring systems:
  • cycloalkyl groups include cyclopropyl, cyclopentyl, cyclohexyl, and
  • halo or “halogen” refers to chloro, bromo, fluoro and iodo.
  • haloalkyl means a substituted alkyl having one or more halo substituents.
  • haloalkyl includes mono, bi, and trifluoromethyl.
  • haloalkoxy means an alkoxy group having one or more halo substituents.
  • haloalkoxy includes OCF 3 .
  • aryl groups include:
  • fluorenyl and the like, which optionally may be substituted at any available carbon or nitrogen atom.
  • a preferred aryl group is optionally-substituted phenyl.
  • heterocycle refers to substituted and unsubstituted 3- to 7-membered monocyclic groups, 7- to 11-membered bicyclic groups, and 10- to 15-membered tricyclic groups, in which at least one of the rings has at least one heteroatom (O, S or N), said heteroatom containing ring preferably having 1, 2, or 3 heteroatoms selected from O, S, and N.
  • Each ring of such a group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less, and further provided that the ring contains at least one carbon atom.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or fully unsaturated.
  • the heterocyclo group may be attached at any available nitrogen or carbon atom.
  • the terms “heterocycle”, “heterocycloalkyl”, “heterocyclo”, “heterocyclic”, and “heterocyclyl” include “heteroaryl” groups, as defined below.
  • exemplary monocyclic heterocyclyl groups include azetidinyl, pyrrolidinyl, oxetanyl, imidazolinyl, oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 1-pyridonyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane and tetrahydro-1,1-dioxothienyl and the like.
  • heteroaryl refers to substituted and unsubstituted aromatic 5- or 6-membered monocyclic groups, 9- or 10-membered bicyclic groups, and 11- to 14-membered tricyclic groups which have at least one heteroatom (O, S or N) in at least one of the rings, said heteroatom-containing ring preferably having 1, 2, or 3 heteroatoms selected from O, S, and N.
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen atoms may optionally be quaternized.
  • Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic.
  • the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. As valence allows, if said further ring is cycloalkyl or heterocyclo it is additionally optionally substituted with ⁇ O (oxo).
  • Exemplary monocyclic heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl and the like.
  • Exemplary bicyclic heteroaryl groups include indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridyl, dihydroisoindolyl, tetrahydroquinolinyl and the like.
  • Exemplary tricyclic heteroaryl groups include carbazolyl, benzindolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • heteroaryl groups include:
  • aryl e.g., phenyl
  • cycloalkyl e.g., cyclohexyl
  • heterocyclo e.g., pyrrolidinyl, piperidinyl, and morpholinyl
  • heteroaryl e.g., tetrazolyl, imidazolyl, pyrazolyl, triazolyl, thiazolyl, and furyl
  • the reference is intended to include rings having 0 to 3, preferably 0 to 2, substituents selected from those recited above for the aryl, cycloalkyl, heterocyclo and/or heteroaryl groups, as appropriate.
  • Carbocyclyl or “carbocyclic” refers to a saturated or unsaturated monocyclic or bicyclic ring in which all atoms of all rings are carbon. Thus, the term includes cycloalkyl and aryl rings.
  • Monocyclic carbocycles have 3 to 6 ring atoms, still more typically 5 or 6 ring atoms.
  • Bicyclic carbocycles have 7 to 12 ring atoms, e.g., arranged as a bicyclo[4,5], [5,5], [5,6] or [6,6] system, or 9 or 10 ring atoms arranged as a bicyclo[5,6] or [6,6] system.
  • Examples of mono- and bicyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, phenyl and naphthyl.
  • the carbocyclic ring may be substituted in which case the substituents are selected from those recited above for cycloalkyl and aryl groups.
  • heteroatoms shall include oxygen, sulfur and nitrogen.
  • the ring or group may be fully unsaturated or partially unsaturated.
  • the compounds of formula I may exist in a free form (with no ionization) or can form salts which are also within the scope of this invention. Unless otherwise indicated, reference to an inventive compound is understood to include reference to the free form and to salts thereof.
  • the term “salt(s)” denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases.
  • the term “salt(s)” may include zwitterions (inner salts), e.g., when a compound of formula I, contains both a basic moiety, such as an amine or a pyridine or imidazole ring, and an acidic moiety, such as a carboxylic acid.
  • Salts of the compounds of the formula I may be formed, for example, by reacting a compound of the formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary acid addition salts include acetates (such as those formed with acetic acid or trihaloacetic acid, for example, trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides (formed with hydrochloric acid), hydrobromides (formed with hydrogen bromide), hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates (formed with maleic acid), methanesulfonates (formed with methanesulf
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts; alkaline earth metal salts such as calcium and magnesium salts; barium, zinc, and aluminum salts; salts with organic bases (for example, organic amines) such as trialkylamines such as triethylamine, procaine, dibenzylamine, N-benzyl- ⁇ -phenethylamine, 1-ephenamine, N,N′-dibenzylethylenediamine, dehydroabietylamine, N-ethylpiperidine, benzylamine, dicyclohexylamine or similar pharmaceutically acceptable amines and salts with amino acids such as arginine, lysine and the like.
  • organic bases for example, organic amines
  • trialkylamines such as triethylamine, procaine, dibenzylamine, N-benzyl- ⁇ -phenethylamine, 1-ephenamine, N,N′-d
  • Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g., decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and others.
  • Preferred salts include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate or nitrate salts.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically-acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically-acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids.
  • the pharmaceutically-acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic,
  • the pharmaceutically-acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th Edition, Mack Publishing Company, Easton, Pa. (1990), the disclosure of which is hereby incorporated by reference.
  • Stereoisomers may include compounds which are optical isomers through possession of one or more chiral atoms, as well as compounds which are optical isomers by virtue of limited rotation about one or more bonds (atropisomers).
  • the definition of compounds according to the invention embraces all the possible stereoisomers and their mixtures. It very particularly embraces the racemic forms and the isolated optical isomers having the specified activity.
  • the racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography.
  • the individual optical isomers can be obtained from the racemates from the conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.
  • the present invention is intended to include all isotopes of atoms occurring in the present compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include deuterium and tritium.
  • Isotopes of carbon include 13 C and 14 C.
  • Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
  • Prodrugs and solvates of the inventive compounds are also contemplated.
  • the term “prodrug” denotes a compound which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of the formula I, and/or a salt and/or solvate thereof. Any compound that will be converted in vivo to provide the bioactive agent (i.e., the compound for formula I) is a prodrug within the scope and spirit of the invention.
  • compounds containing a carboxy group can form physiologically hydrolyzable esters which serve as prodrugs by being hydrolyzed in the body to yield formula I compounds per se.
  • Such prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes.
  • esters of compounds of formula I include C 1-6 alkylbenzyl, 4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, C 1-6 alkanoyloxy-C 1-6 alkyl, e.g., acetoxymethyl, pivaloyloxymethyl or propionyloxymethyl, C 1-6 alkoxycarbonyloxy-C 1-6 alkyl, e.g., methoxycarbonyl-oxymethyl or ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyl-2-oxo-1,3-dioxolen-4-yl)-methyl and other well known physiologically hydrolyzable esters used, for example, in the penicillin and cephalosporin arts. Such esters may be prepared by conventional techniques known in the art.
  • prodrug derivatives are well known in the art.
  • prodrug derivatives see:
  • solvates e.g., hydrates
  • Methods of solvation are generally known in the art.
  • the compounds of the invention modulate IL-23-stimulated and IFN ⁇ -stimulated cellular functions, including gene transcription.
  • Other types of cellular functions that may be modulated by the compounds of the instant invention include, but are not limited to, IL-12-stimulated responses.
  • compounds of formula I have utility in treating conditions associated with the modulation of the function of IL-23 or IFN ⁇ , and particularly the selective inhibition of function of IL-23, IL-12 and/or IFN ⁇ , by acting on Tyk2 to mediate signal transduction.
  • Such conditions include IL-23-, IL-12-, or IFN ⁇ -associated diseases in which pathogenic mechanisms are mediated by these cytokines.
  • the terms “treating” or “treatment” encompass the treatment of a disease state in a mammal, particularly in a human, and include: (a) preventing or delaying the occurrence of the disease state in a mammal, in particular, when such mammal is predisposed to the disease state but has not yet been diagnosed as having it; (b) inhibiting the disease state, i.e., arresting its development; and/or (c) achieving a full or partial reduction of the symptoms or disease state, and/or alleviating, ameliorating, lessening, or curing the disease or disorder and/or its symptoms.
  • compounds of Formula I are useful in treating IL-23-, IL-12- or IFN ⁇ -associated diseases including, but not limited to, inflammatory diseases such as Crohn's disease, ulcerative colitis, asthma, graft versus host disease, allograft rejection, chronic obstructive pulmonary disease; autoimmune diseases such as Graves' disease, rheumatoid arthritis, systemic lupus erythematosis, cutaneous lupus, lupus nephritis, discoid lupus erythematosus, psoriasis; auto-inflammatory diseases including CAPS, TRAPS, FMF, adult onset stills, systemic onset juvenile idiopathic arthritis, gout, gouty arthritis; metabolic diseases including type 2 diabetes, atherosclerosis, myocardial infarction; destructive bone disorders such as bone resorption disease
  • the specific conditions or diseases that may be treated with the inventive compounds include, without limitation, pancreatitis (acute or chronic), asthma, allergies, adult respiratory distress syndrome, chronic obstructive pulmonary disease, glomerulonephritis, rheumatoid arthritis, systemic lupus erythematosis, cutaneous lupus, lupus nephritis, discoid lupus erythematosus, scleroderma, chronic thyroiditis, Graves' disease, autoimmune gastritis, diabetes, autoimmune hemolytic anemia, autoimmune neutropenia, thrombocytopenia, atopic dermatitis, chronic active hepatitis, myasthenia gravis, multiple sclerosis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, psoriasis, graft vs.
  • Preferred methods of treatment are those wherein the condition is selected from Crohn's disease, ulcerative colitis, allograft rejection, rheumatoid arthritis, psoriasis, ankylosing spondylitis, psoriatic arthritis, and pemphigus vulgaris.
  • preferred methods of treatment are those wherein the condition is selected from ischemia reperfusion injury, including cerebral ischemia reperfusions injury arising from stroke and cardiac ischemia reperfusion injury arising from myocardial infarction.
  • Another preferred method of treatment is one in which the condition is multiple myeloma.
  • IL-23-, IL-12- and/or IFN ⁇ -associated condition or “IL-23-, IL-12- and/or IFN ⁇ -associated disease or disorder” are used herein, each is intended to encompass all of the conditions identified above as if repeated at length, as well as any other condition that is affected by IL-23, IL-12 and/or IFN ⁇ .
  • the present invention thus provides methods for treating such conditions, comprising administering to a subject in need thereof a therapeutically-effective amount of at least one compound of Formula I or a salt thereof “Therapeutically effective amount” is intended to include an amount of a compound of the present invention that is effective when administered alone or in combination to inhibit IL-23, IL-12 and/or IFN ⁇ function and/or treat diseases.
  • the methods of treating IL-23-, IL-12 and/or IFN ⁇ -associated conditions may comprise administering compounds of Formula I alone or in combination with each other and/or other suitable therapeutic agents useful in treating such conditions.
  • therapeutically effective amount is also intended to include an amount of the combination of compounds claimed that is effective to inhibit IL-23, IL-12 and/or IFN ⁇ function and/or treat diseases associated with IL-23, IL-12 and/or IFN ⁇ .
  • Such other therapeutic agents include corticosteroids, rolipram, calphostin, cytokine-suppressive anti-inflammatory drugs (CSAIDs), Interleukin-10, glucocorticoids, salicylates, nitric oxide, and other immunosuppressants; nuclear translocation inhibitors, such as deoxyspergualin (DSG); non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen, celecoxib and rofecoxib; steroids such as prednisone or dexamethasone; antiviral agents such as abacavir; antiproliferative agents such as methotrexate, leflunomide, FK506 (tacrolimus, PROGRAF®); anti-malarials such as hydroxychloroquine; cytotoxic drugs such as azathiprine and cyclophosphamide; TNF- ⁇ inhibitors such as tenidap, anti-TNF antibodies or soluble TNF receptor,
  • the above other therapeutic agents when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • PDR Physicians' Desk Reference
  • such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the inventive compounds.
  • the present invention also provides pharmaceutical compositions capable of treating IL-23-, IL-12- or IFN ⁇ -associated conditions by inhibiting Tyk2-mediated signal transduction, including IL-23-, IL-12- and/or IFN ⁇ -mediated diseases, as described above.
  • inventive compositions may contain other therapeutic agents as described above and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (e.g., excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulation.
  • pharmaceutical additives e.g., excipients, binders, preservatives, stabilizers, flavors, etc.
  • the present invention further includes compositions comprising one or more compounds of Formula I and a pharmaceutically acceptable carrier.
  • a “pharmaceutically acceptable carrier” refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals.
  • Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include without limitation the type and nature of the active agent being formulated; the subject to which the agent-containing composition is to be administered; the intended route of administration of the composition; and, the therapeutic indication being targeted.
  • Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms.
  • Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art.
  • suitable pharmaceutically acceptable carriers, and factors involved in their selection are found in a variety of readily available sources such as, for example, Remington's Pharmaceutical Sciences, 17th Edition (1985), which is incorporated herein by reference in its entirety.
  • the compounds of Formula I may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or quantity of drug to be delivered.
  • Topical administration is generally preferred for skin-related diseases, and systematic treatment preferred for cancerous or pre-cancerous conditions, although other modes of delivery are contemplated.
  • the compounds may be delivered orally, such as in the form of tablets, capsules, granules, powders, or liquid formulations including syrups; topically, such as in the form of solutions, suspensions, gels or ointments; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular or intrasternal injection or infusion techniques (e.g., as sterile injectable aq. or non-aq.
  • Dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents may be administered.
  • the compounds may be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved with suitable pharmaceutical compositions or, particularly in the case of extended release, with devices such as subcutaneous implants or osmotic pumps.
  • compositions for topical administration include a topical carrier such as PLASTIBASE® (mineral oil gelled with polyethylene).
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the inventive compounds may also be orally delivered by sublingual and/or buccal administration, e.g., with molded, compressed, or freeze-dried tablets.
  • compositions may include fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • high molecular weight excipients such as celluloses (AVICEL®) or polyethylene glycols (PEG); an excipient to aid mucosal adhesion such as hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), sodium carboxymethyl cellulose (SCMC), and/or maleic anhydride copolymer (e.g., GANTREZ®); and agents to control release such as polyacrylic copolymer (e.g., CARBOPOL 934®).
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • compositions for nasal aerosol or inhalation administration include solutions which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance absorption and/or bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • compositions for rectal administration include suppositories which may contain, for example, suitable non-irritating excipients, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • suitable non-irritating excipients such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • the therapeutically-effective amount of a compound of the present invention may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a mammal of from about 0.05 to 1000 mg/kg; 1-1000 mg/kg; 1-50 mg/kg; 5-250 mg/kg; 250-1000 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day.
  • the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats, horses, and the like.
  • this term is intended to include all subjects, most preferably mammalian species that are affected by modulation of IL-23, IL-12 and/or IFN ⁇ -mediated functions.
  • the probe displacement assay is conducted as follows: In a 385 well plate, test compounds along with recombinantly expressed His-tagged protein corresponding to amino acids 575-869 of human Tyk2 (sequence shown below) at 2.5 nM, 40 nM ((R)—N-(1-(3-(8-methyl-5-(methylamino)-8H-imidazo[4,5-d]thiazolo[5,4-b]pyridin-2-yl)phenyl)ethyl)-2-([ 3 H]methylsulfonyl)benzamide) (preparation described below) and 80 ⁇ g/mL Copper His-Tag scintillation proximity assay beads (Perkin Elmer, Catalog #RPNQ0095) in 50 mM HEPES, pH 7.5, containing 100 ⁇ g/mL bovine serum albumin and 5% DMSO were incubated for 30 minutes at room temperature.
  • the amount of radiolabeled probe (preparation described below) bound to Tyk2 was then quantified by scintillation counting, and the inhibition by the test compound calculated by comparison to wells either with no inhibitor (0% inhibition) or without Tyk2 (100% inhibition).
  • the IC 50 value is defined as the concentration of test compound required to inhibit radiolabeled probe binding by 50%.
  • radiolabeled probe (R)—N-(1-(3-(8-methyl-5-(methylamino)-8H-imidazo[4,5-d]thiazolo[5,4-b]pyridin-2-yl)phenyl)ethyl)-2-([ 3 H]methylsulfonyl)benzamide, was performed as described below.
  • 2-([ 3 H]Methylsulfonyl)benzoic acid 2-Mercaptobenzoic acid (2.3 mg, 0.015 mmol) and cesium carbonate (2 mg, 0.006 mmol) were added to a 5 mL round-bottomed flask. The flask was attached to a ported glass vacuum line and anhydrous DMF (0.5 mL) was introduced with magnetic stirring. An ampoule of tritiated methyl iodide (200 mCi, Perkin-Elmer lot 3643419) was added to the reaction flask and stirring was maintained at rt for 3 h. In-process HPLC analysis with radiometric detection indicated 80% conversion to the desired product by comparison with authentic standard.
  • the radiochemical purity was measured by HPLC to be 99% (Luna 5 ⁇ , C18 (4.6 ⁇ 150 cm); A: H 2 O (0.1% TFA); B: MeOH; 1.2 ml/min; 270 nm; 0-10 min 20% B; 10-15 min 20-100% B; 15-25 min 100% B.
  • the product was dissolved in anhydrous acetonitrile to give a final solution activity of 5.8 mCi/mL.
  • the purification routine was performed a second time to yield a total of 1.7 mCi (7% radiochemical yield) of the desired product in 99.9% radiochemical purity.
  • Mass spectral analysis of the tritiated product (m/z M+H 527.33) was used to establish the specific activity at 80.6 Ci/mmol.
  • Probe Displacement Data Probe Displacement Example No. (EC 50 , ⁇ M) 4 0.13 5 0.41 10 0.10 16 6.57E ⁇ 03 51 7.19E ⁇ 03 52 5.13E ⁇ 03 61 1.66E ⁇ 03 67 6.53E ⁇ 03 69 0.07 70 5.22E ⁇ 03 73 5.21E ⁇ 03 75 6.18E ⁇ 03 76 6.17E ⁇ 03 84 1.28E ⁇ 03 85 7.36E ⁇ 03 87 2.02E ⁇ 03 94 1.72E ⁇ 03 102 1.59E ⁇ 03 108 1.46E ⁇ 03 112 1.94E ⁇ 03 114 1.89E ⁇ 03 125 0.11 134 5.64E ⁇ 03 140 0.07 142 6.95E ⁇ 03 146 1.70E ⁇ 03 147 8.77E ⁇ 04 151 7.22E ⁇ 03 154 0.09 155 7.13E ⁇ 03 160 0.07 176 6.35E ⁇ 03 181 6.97E ⁇ 03 183 5.72E ⁇ 03 186 0.06 188 5.10E ⁇ 03 194 0.08 Kit225 T Cell Assay
  • Kit225 T cells with a stably-integrated STAT-dependent luciferase reporter were plated in RPMI (Gibco) containing 10% heat-inactivated FBS (Gibco) and 100 U/mL PenStrep (Gibco). The cells were then stimulated with either 20 ng/mL human recombinant IL-23 or 200 U/mL human recombinant IFN ⁇ (PBL InterferonSource) for 5-6 hours. Luciferase expression was measured using the STEADYGLO® Luciferase Assay System (Promega) according to the manufacturer's instructions. Inhibition data were calculated by comparison to no inhibitor control wells for 0% inhibition and non-stimulated control wells for 100% inhibition. Dose response curves were generated to determine the concentration required to inhibit 50% of cellular response (IC 50 ) as derived by non-linear regression analysis.
  • the compounds of the present invention may be synthesized by many methods available to those skilled in the art of organic chemistry.
  • General synthetic schemes for preparing compounds of the present invention are described below. These schemes are illustrative and are not meant to limit the possible techniques one skilled in the art may use to prepare the compounds disclosed herein. Different methods to prepare the compounds of the present invention will be evident to those skilled in the art. Additionally, the various steps in the synthesis may be performed in an alternate sequence in order to give the desired compound or compounds. Examples of compounds of the present invention prepared by methods described in the general schemes are given in the preparations and examples section set out hereinafter.
  • Scheme 1 illustrates the preparation of title compounds of the invention (I) from the intermediate pyridazine (II) or 1,2,4-triazine (III) along with an amine (IV).
  • the coupling of the halo-pyridazine may be affected by many of the ways known to achieve displacement of 6-halo-pyridazines by amines. This includes, but is not limited to, the palladium catalyzed N-arylation of amines, and nucleophilic displacement of the halide by the amine.
  • palladium sources can be used to affect the coupling including both palladium(II) salts (for example palladium diacetate) as well as neutral palladium (such as tetrakis triphenylphosphine palladium or tris(dibenzylideneacetone)dipalladium).
  • palladium(II) salts for example palladium diacetate
  • neutral palladium such as tetrakis triphenylphosphine palladium or tris(dibenzylideneacetone)dipalladium.
  • catalyst ligands are suitable for this transformation including bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos) and 2-(dicyclohexylphosphino)-3,6-dimethoxy-2′,4′,6′-tri-i-propyl-1,1′-biphenyl (BrettPhos) and many others that those versed in synthetic chemistry are familiar with (see Surry, D. S. et al., Chem. Sci., 2:27-50 (2011)).
  • a variety of bases can be employed (such as potassium carbonate, sodium tert-butoxide, cesium carbonate and the like) as well as a number of solvents (such as 1,4-dioxane, toluene and dimethylacetamide and the like).
  • Nucleophilic displacement is generally possible at elevated temperatures (typically >100° C.) in the presence or absence of either an acid or base catalyst. Heating can be accomplished using either a microwave or conventional heating. Amines are most typically, but not exclusively, aliphatic in such displacements.
  • Scheme 2 illustrates the preparation of the amides II/III from the corresponding carboxylic acids (V/VI) by coupling with an amine (VII).
  • This coupling may be affected by many of the ways known to prepare carboxamides.
  • condensation of acid with amine (III) (VII) may be effected by treatment of the carboxylic acid with an activating reagent, such as a water-soluble carbodiimide (EDC), in the presence of an N-hydroxy triazole (HOAt or HOBt, or the like) and amine (III) (VII) in the presence of base (preferably triethylamine, diisopropylethylamine, or the like) in an appropriate polar aprotic solvent (N,N-dimethylformamide, acetonitrile, dichloromethane, or the like).
  • an activating reagent such as a water-soluble carbodiimide (EDC)
  • HOAt or HOBt N-hydroxy triazole
  • base
  • the carboxylic acid may also be converted to an acid chloride by treatment with an appropriate chlorinating agent (thionyl chloride, oxalyl chloride, or the like).
  • the carboxylic acid may be converted to an acyl fluoride upon exposure to a fluorinating agent (such as cyanuric fluoride).
  • a fluorinating agent such as cyanuric fluoride
  • Condensation of the acyl halide (chloride or fluoride) with the amine III (VII) may then provide the amide II/III.
  • Scheme 3 illustrates the preparation of acids V/VI via saponification of ester VIII/IX. Saponification can be accomplished using sodium, lithium, or potassium hydroxide under aqueous conditions with an organic co-solvent such as methanol and/or tetrahydrofuran.
  • an organic co-solvent such as methanol and/or tetrahydrofuran.
  • Scheme 4 illustrates the preparation of VIII/IX from the chloro-heterocycles VXI X/XI via coupling with an amine (XII).
  • this coupling can be accomplished using nucleophilic displacement, using either strong bases (for example lithium hexamethyldisilyazide hexamethyldisilazide) or weak bases (for example triethylamine) in an appropriate solvent (tetrahydrofuran, acetonitrile, dimethylformamide and related). Careful monitoring of the reactions progress and appropriate solvent/base selection ensure that regioselectivity and over addition are not a concern.
  • strong bases for example lithium hexamethyldisilyazide hexamethyldisilazide
  • weak bases for example triethylamine
  • Scheme 5 illustrates the preparation of X, which was carried out in the manner previously described in US 2004/0142930 A1 (see: Yamada, K. et al., “Preparation of Heterocyclic Compounds as Selective Phosphodiesterase V Inhibitors”, US 2004/0142930 A1 (Jul. 22, 2004)).
  • Scheme 7 illustrates an alternative preparation of II.
  • the amine XII is coupled to the dichloride XVII.
  • Displacement of the dihalide is most often accomplished in the presence of a strong base, such as sodium bis(trimethylsilyl)amide or lithium bis(trimethylsilyl)amide, but it is also conceivable that it could be accomplished using a weak base such as N,N-diisopropylethylamine (or related), or under elevated thermal conditions in the absence of any base, or in the presence of an acid catalyst.
  • a number of solvents could be used, including tetrahydrofuran, dimethylformamide and N-methyl-2-pyrrolidone.
  • Scheme 8 illustrates the preparation of XI, which may be carried out in the manner previously described in US 2002/0061865 A1 (see: Kramer, J. B. et al., “Pyridotriazines and Pyridopyridazines”, US 2002/0061865 A1 (May 23, 2002).).
  • Scheme 9 illustrates how pendant sulfides can be oxidized to the corresponding sulfones or (in the case of XXII) the sulfoxide (not illustrated).
  • the sulfide (XXII/XXIII) can be oxidized to the sulfone (XXIV/XXV) using an oxidant such as sodium tungstate or 3-chloroperbenzoic acid in an organic solvent such as dichloromethane or acetic acid.
  • the partial oxidation of XXII to the sulfoxide generally requires more mild conditions such as hydrogen peroxide in acetic acid; however, it is possible to use the same conditions as when targeting the sulfone if one quenches the reaction at the appropriate time.
  • the sulfide group (Z) can be displaced by VII (Scheme 2) and then partial oxidation can be performed as described above.
  • the Williamson ether formation is a common protocol for the synthesis of ethers, the reaction consists of the combination of an alcohol and a base-such as potassium carbonate, sodium hydride, triethylamine, or any number of others, followed by the addition of a compatible electrophile, such as an aliphatic, benzylic or allylic functional group featuring a leaving group-most commonly a halide, but mesylates/tosylates and other groups are also compatible, is added.
  • a compatible electrophile such as an aliphatic, benzylic or allylic functional group featuring a leaving group-most commonly a halide, but mesylates/tosylates and other groups are also compatible.
  • the reaction is typically run in a polar aprotic solvent such as tetrahydrofuran or dimethylformamide.
  • nitro group of XXVII is then reduced to the amine (XXVIII) using a heterogeneous catalyst such as palladium, zinc or iron and a hydrogen source such as hydrogen (gas), ammonium chloride or hydrochloric acid, such reactions are typically run in alcoholic solvents.
  • a heterogeneous catalyst such as palladium, zinc or iron and a hydrogen source such as hydrogen (gas), ammonium chloride or hydrochloric acid
  • the boronic ester (XXIX) can be coupled via the Suzuki coupling to a wide variety of aryl and heteroaryl halides using a number of different catalysts, ligands, bases and solvents.
  • One common combination of reagents is 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride, as the catalyst, tribasic potassium phosphate (in water), as the base, reacting with an aryl bromide using dioxane as the solvent; however, a great number of potential combinations exist, for a partial description see: Barder, T. E. et al., J. Am. Chem. Soc., 127:4685-4696 (2005); and Miyaura, N. et al., Chem. Rev., 95:2457-2483 (1995).
  • Scheme 11 illustrates a means by which diversity at the R 7 (Ia) can be introduced at the end of the synthetic sequence.
  • XVII and XXVIII can be coupled following the same procedures described in Scheme 7.
  • Intermediate XXX can be converted to the primary amine via the addition of a protected amine (either via thermal, or selective palladium catalyzed N-arylation conditions) followed by deprotection, for example 4-methoxyphenyl)methanamine can be introduced under strictly thermal conditions followed by deprotection with a protic acid (such as trifluoroacetic acid) to provide XXXI.
  • a protic acid such as trifluoroacetic acid
  • Scheme 12 illustrates how some of heterocycles can be built directly off of carbonyl functionality to arrive at anilines XII without the use of a transition metal catalyzed coupling reaction.
  • the commercially available XXXIV can be converted to the ether XXXV via the techniques described in Scheme 10, similarly XXXVI can be converted to XXXVII.
  • XXXV can be converted to the amide XXXVIII directly using ammonia and ammonium hydroxide in methanol, or via saponification and amide formation (described in Schemes 3 and 2 respectively).
  • the amide XXXVIII can be converted to a triazole via formation of the amidine using reagents such as N,N-dimethylacetamide dimethyl acetal or N,N-dimethylformamide dimethyl acetal followed by exposure to hydrazine in the presence of acetic acid.
  • reagents such as N,N-dimethylacetamide dimethyl acetal or N,N-dimethylformamide dimethyl acetal followed by exposure to hydrazine in the presence of acetic acid.
  • the tetrazole XL can be prepared from XXXVIII by reaction with triazidochlorosilane (generated in situ from tetrachlorosilane and sodium azide, see: El-Ahl, A-A. S. et al., Tetrahedron Lett., 38:1257-1260 (1997).).
  • the hydrazide XLI can be converted to the oxadiazole via a condensation reaction with an orthoformate or orthoacetate under thermal or acid catalyzed conditions, often using the orthoformate/orthoacetate as the solvent.
  • the aceto variant of hydrazide XLI can be converted to the thiazole by exposure to a sulfonating reagent such as Lawesson's reagent and then condensation under thermal conditions, typically in polar aprotic solvent such as dioxane.
  • the ketone XXXVII can be converted to the pyrazole XLIV by condensation with N,N-dimethylacetamide dimethyl acetal or N,N-dimethylformamide dimethyl acetal (or related) followed by reaction with hydrazine in the presence of acetic acid.
  • the heterocycle can further be reacted with an electrophile such as organo-halides, epoxides or activated carbonyl species (under basic conditions using an inorganic base such as potassium carbonate, a tertiary amine such as triethylamine, or a strong base such as sodium hydride) or with vinyl ethers such as ethoxyethene (under acidic conditions).
  • an electrophile such as organo-halides, epoxides or activated carbonyl species (under basic conditions using an inorganic base such as potassium carbonate, a tertiary amine such as triethylamine, or a strong base such as sodium hydride) or with vinyl ethers such as ethoxyethene (under acidic conditions).
  • Other electrophiles such as silyl halides would also be successful as would potentially a selective palladium catalyzed N-arylation.
  • the nitro compounds can be converted to the aniline XII via
  • Scheme 13 illustrates the synthesis of the thio-variant of XII.
  • XLVI which can be converted to the ester via heating with methanol in the presence of a protic acid, as well as by any number of techniques available for the synthesis of esters from acids, such as formation of the acid halide (described in Scheme 2) followed by reaction with methanol.
  • Displacement of the chloride to provide XLVIII can be accomplished via nucleophilic addition using sodium thiomethoxide.
  • Conversion to the functionalized aniline XLIX follows the same techniques illustrated and described in Scheme 12. Additionally the final sulfide product can be oxidized to the sulfone using the oxidation conditions described in Scheme 9.
  • Scheme 14 illustrates another form of the final compound Ia.
  • aniline L made via reduction of the nitro compound XXXV by analogy to Scheme 10
  • dichloride XVII using the techniques from Scheme 7.
  • Conversion to LII can be accomplished using the same techniques described in Scheme 1.
  • Saponification (described in Scheme 3) provides the acid LIII.
  • the acid LIII can be converted to various heterocycles using the techniques described in Scheme 12, or it can be coupled with an amine to generate the amide LV as the final product as described in Scheme 2.
  • Scheme 15 illustrates another variant of XII, where the aniline has been substituted with a heterocycle via a carbon-nitrogen bond.
  • an Ullmann condensation for a recent review see: Mannier, F. et al., Angew. Chem. Int. Ed., 48:6954-6971 (2009)
  • This reaction is typically performed in the presence of a copper salt (such as copper(I) oxide), an inorganic base (such as cesium carbonate) and often a ligand (although some solvents such as DMF can take the role of the ligand).
  • the phenol LVI can be converted to the ether LVII using the Williamson ether conditions as described in Scheme 10. Conversion to the aniline (LVIII) is accomplished by reduction of the nitro group as described in Scheme 10.
  • Scheme 16 describes the synthesis of anilines LIX and LXII.
  • a Sonogashira coupling of XXVIII/XXVII with ethynyltrimethylsilane followed by removal of the silyl group using a mild base (such as potassium carbonate in a protic solvent such as methanol) or a fluoride source (such as tetrabutylammonium fluoride or potassium fluoride) can be used to provide the terminal alkynes LIX and LX.
  • the Sonogashira coupling is performed using a palladium catalyst (such as tetrakis triphenylphosphine palladium), a copper catalyst such as copper(I) iodide, and a base (typically an amine base such as triethylamine or diisopropylamine) using either the base as the solvent or a polar solvent such as dimethylformamide; however, a great deal of work has been done running the reaction with different ligands and additives and even in the absence of the catalysts, see: Chinchilla, R. et al., Chem. Rev. 107:874-923 (2007); Chinchilla, R. et al., Chem. Soc.
  • a palladium catalyst such as tetrakis triphenylphosphine palladium
  • a copper catalyst such as copper(I) iodide
  • a base typically an amine base such as triethylamine or diisopropylamine
  • the aniline LIX can be coupled to XVII as described in Scheme 7 and then converted to the target ligand I as described in Scheme 1 or further elaborated using the techniques described for LXI (to follow).
  • LX can be converted to the 1,2,3-triazole using the Huisgen cycloaddition (or “Click chemistry”), This reaction is run between an alkyne and an azide using a copper catalyst (commonly copper(II) sulfate), a reducing agent (such as sodium ascorbate), the reaction can be run in a number of solvents/co-solvents including water, tert-butyl alcohol, tetrahydrofuran and toluene.
  • a copper catalyst commonly copper(II) sulfate
  • a reducing agent such as sodium ascorbate
  • Scheme 17 illustrates the synthesis of penultimate compounds LXV (converted to target ligands using the coupling procedures described in Scheme 1).
  • Intermediate LXIII (prepared using the techniques described in Scheme 16 and Scheme 7) can be converted to the isoxazole LXV using a [3+2] cycloaddition with a nitrile oxide (formed in situ from a N-hydroxyimidoyl chloride and a mild non-nucleophilic base).
  • the reaction can be run thermally in aprotic solvents (such as dichloroethane) but recent work has described the utility of catalysts in the reaction, see: napn, S. et al., Angew. Chem. Int. Ed., 47:8285-8287 (2008).
  • Scheme 18 illustrates the synthesis of target compounds LXX and LXXI.
  • Commercially available LXVI can be converted to the aniline LXVIII following the strategies outlined in Scheme 10. Addition of LXVIII to XVII follows the techniques described in Scheme 7 to provide LXIX which can be coupled to amines IV following the strategies described in Scheme 1.
  • Conversion of the cyano-containing LXX to the oxadiazole LXXI can be accomplished via the nucleophilic addition of hydroxylamine to the cyanide, performed under basic conditions typically in a polar protic solvent such as water or alcohol, followed by acylation and condensation with acetic anhydride, done by heating the intermediate with acetic anhydride in a polar aprotic solvent such as dioxane.
  • a polar protic solvent such as water or alcohol
  • Preparation of compounds of Formula (I), and intermediates used in the preparation of compounds of Formula (I), can be prepared using procedures shown in the following Examples and related procedures. The methods and conditions used in these examples, and the actual compounds prepared in these Examples, are not meant to be limiting, but are meant to demonstrate how the compounds of Formula (I) can be prepared. Starting materials and reagents used in these examples, when not prepared by a procedure described herein, are generally either commercially available, or are reported in the chemical literature, or may be prepared by using procedures described in the chemical literature.
  • the phrase “dried and concentrated” generally refers to drying of a solution in an organic solvent over either sodium sulfate or magnesium sulfate, followed by filtration and removal of the solvent from the filtrate (generally under reduced pressure and at a temperature suitable to the stability of the material being prepared).
  • Column chromatography was performed with pre-packed silica gel cartridges using an Isco medium pressure chromatography apparatus (Teledyne Corporation), eluting with the solvent or solvent mixture indicated.
  • Chemical names were determined using ChemDraw Ultra, version 9.0.5 (CambridgeSoft). The following abbreviations are used:
  • Reverse-phase preparative high performance liquid chromatography (“HPLC”) was performed with Shimadzu 8A liquid chromatographs using YMC S5 ODS columns (20 ⁇ 100, 20 ⁇ 250, or 30 ⁇ 250 millimeter (“mm”)). Gradient elution was performed with methanol (“MeOH”)/water mixtures in the presence of 0.1% trifluoroacetic acid (“TFA”).
  • HPLC high performance liquid chromatography
  • UV Ultraviolet
  • Solvent A 0.2% phosphoric acid, 90% water, 10% methanol
  • Solvent B 0.2% phosphoric acid, 90% methanol, 10% water
  • Int1 (12.2 g, 50.8 mmol) was dissolved in diethyl ether (100 mL) and triphenylphosphine (14 g, 53.5 mmol) was added. The reaction was stirred overnight at room temperature and then concentrated in vacuo. To the residual sludge was added acetic acid (100 mL) and water (10 mL), the vessel was equipped with a condenser and heated to reflux for 6 hours, and then concentrated in vacuo. The crude sludge was purified by automated chromatography (DCM/MeOH) and then by titration with diethyl ether ( ⁇ 2) to provide Int2 (5.25 g, 28.5 mmol).
  • DCM/MeOH automated chromatography
  • ⁇ 2 diethyl ether
  • Int4 (65 mg, 0.20 mmol) was dissolved in tetrahydrofuran (THF, 2 mL) and lithium hydroxide (2 M in water, 0.40 mL, 0.80 mmol) was added. After stirring 30 min at room temperature, the THF was removed under reduced pressure. The residual solution was diluted with water and then acidified with 1 M hydrochloric acid. The product was extracted three times with ethyl acetate and then the combined organic layers were dried over sodium sulfate, filtered and concentrated.
  • THF tetrahydrofuran
  • Int5 can be prepared as follows:
  • Int1 (41.6 g, 182 mmol) was dissolved in diethyl ether (300 mL) and triphenylphosphine (47.8 g, 182 mmol) was added. The reaction was stirred overnight at room temperature and then concentrated in vacuo. To the residual sludge was added acetic acid (300 mL) and water (30 mL), the vessel was equipped with a condenser and heated to reflux for 6 hours. The reaction was concentrated and then dissolved in 1,2-dichloroethane (300 mL) and re-concentrated. The resultant slurry was dissolved in THF (600 mL) and MeOH (200 mL) and then LiOH (3M aq.
  • Int7 (10 g, 64.1 mmol) was placed in a 1 L RBF and triethylamine (8.9 mL, 64.1 mmol) was added, followed by phosphorus oxychloride (50 mL, 546 mmol).
  • the flask was placed in a room temperature oil bath and once self-reflux ceased, the temperature was raised to 80° C. Once that temperature was reached and the vigorous reflux subsided the temperature was raised again to 110° C. and the reaction run for 120 minutes. The heating was stopped and the reaction allowed to cool to ⁇ 90° C.
  • the crude product was diluted with DMF and filtered, and then purified using preparative HPLC.
  • the pure fractions were pooled and concentrated in vacuo to a volume of about 2 mL at which point saturated aqueous sodium bicarbonate was added and the slurry stirred for 10 minutes.
  • the product was extracted with ethyl acetate ( ⁇ 5), the combined organic layers were washed with deionized water, dried over sodium sulfate, filtered and concentrated.
  • the residual solid was dissolved in 2:1 acetonitrile:water, frozen and then dried on a lyopholizer overnight to provide 1 (8.4 mg, 0.019 mmol).
  • Example 2 The following Examples were prepared in a similar manner to the product of Example 1:
  • Step 1 The crude product of Step 1 was dissolved in DMF (3 mL), potassium carbonate (269 mg, 2.0 mmol) was added and the reaction was stirred for 30 minutes. Next iodomethane (0.12 mL, 2.0 mmol) was added and the reaction was stirred for 2 hours. The crude product was filtered, concentrated and purified by automated chromatography providing 1-(2-methoxy-3-nitrophenyl)-1H-pyrazole (115 mg, 39% yield). LC retention time 1.34 [J].
  • the vessel was then sealed and placed into a warm 105° C. bath. Stirred at 105° C. overnight. After stirring overnight, evaporated away the diisopropylamine and the excess TMS-acetylene, then diluted with 150 mL ethyl acetate. Washed the organic solution once with 1:1 ammonium hydroxide:sat. ammonium chloride, once with saturated ammonium chloride, once with 10% aqueous LiCl, and once with brine. The organic layer was then dried over sodium sulfate, filtered, concentrated, and loaded onto a 24 g silica gel column for purification by flash chromatography, eluting with 0-100% EtOAc in hexanes. Afforded ((2-methoxy-3-nitrophenyl)ethynyl)trimethylsilane (177 mg, 28% yield) as an impure brown oil.
  • the resultant oil was loaded onto a 12 g silica gel column, then purified by flash chromatography, eluting with 0-10% MeOH in dichloromethane. Afforded 1-ethynyl-2-methoxy-3-nitrobenzene (74 mg, 0.397 mmol, 55.9% yield) as a brown oil.
  • Benzoic acid (2 mg, 0.016 mmol), L-ascorbic acid sodium salt (2 mg, 10.10 ⁇ mol), and copper(II) sulfate (2 mg, 0.013 mmol) were all weighed into the small flask containing 1-ethynyl-2-methoxy-3-nitrobenzene (74 mg, 0.418 mmol).
  • Isomer B A mixture of 4-(2-methoxy-3-nitrophenyl)-1-methyl-1H-1,2,3-triazole (21 mg, 0.09 mmol), zinc (58.6 mg, 0.897 mmol) and ammonium chloride (48 mg, 0.897 mmol) in EtOH (1 mL) and water (0.143 mL) was stirred at room temperature for 1 hr. The reaction was then diluted with dichloromethane (50 ml), and filtered.
  • Methyl 2-methoxy-3-nitrobenzoate (11 g, 52.1 mmol) was dissolved in a cold solution of ammonia in methanol (7N, 250 mL) and conc. aqueous ammonium hydroxide (100 mL) was added. The flask was sealed and the resulting solution was allowed to gently stir at room temperature overnight ( ⁇ 17 h). The reaction mixture was concentrated on the rotovap using a slightly warm water bath to yield an aqueous slurry of the product. This slurry was diluted with additional water ( ⁇ 300 mL) and was sonicated briefly then the solid was collected by vacuum filtration and the resulting yellow solid was rinsed with additional water ( ⁇ 100 mL).
  • Trifluoroacetic acid (0.787 mL, 10.60 mmol) was added to a stirred solution of tert-butyl 2-(2-methoxy-3-nitrobenzoyl) hydrazinecarboxylate (1.10 g, 3.53 mmol) in dichloromethane (10 mL) at room temperature. The reaction mixture was stirred for one hour at room temperature. The reaction mixture was concentrated under vacuum with repeated additions of dichloromethane to evaporate of residual TFA to give 0.730 g tan solid product. (Yield 98%). LC retention time 0.70 [A]. MS(E + ) m/z: 212 (MH + ).
  • N′-acetyl-2-methoxy-3-nitrobenzohydrazide 500 mg, 1.975 mmol
  • dioxane 20 mL
  • Lawesson's reagent 2.00 g, 4.94 mmol
  • the reaction was then cooled to room temperature and concentrated and partitioned between water and ethyl acetate. The two layers were separated and the aqueous layer extracted three times with ethyl acetate. The combined organic layers were washed with 10% sodium bicarbonate solution followed by brine.
  • the reaction was concentrated on a rotary evaporator until the majority of the THF was removed and a precipitate prevailed throughout the vessel. Water ( ⁇ 500 mL) was then added and the slurry sonicated for 5 minutes and stirred for 15 min. The solid was filtered off, rinsing with water and then air dried for 30 minutes. The powder was collected and dissolved in dichloromethane. The organic layer was washed with water and brine and then dried over sodium sulfate, filtered and concentrated to provide the product (12.5 g, 66% yield) (carried on as is).
  • Example 52 To a homogeneous solution of Example 52 (50 mg, 0.12 mmol) in dichloromethane (3 mL) was added HCl (1M aq., 0.13 mL, 0.13 mmol) resulting in the solution turning yellow. The homogenous solution was concentrated down and then re-concentrated from dichloromethane twice to remove residual water, resulting in a white powder. The powder was suspended in dichloromethane and sonicated for 15 minutes, the powder was then collected via filtration, rinsing with dichloromethane to provide the corresponding HCl salt (38 mg, 70% yield).
  • Example 52 The following Examples were prepared in a similar manner to the product of Example 52.
  • the aniline used in each case was prepared following the preparation number, or in a manner similar to it, as denoted for each entry:
  • Example 164 The following Examples were prepared in a similar manner to the product of Example 164:
  • Int21 (prepared in a similar manner to Example 164) (30 mg, 0.076 mmol) was slurried in N,N-dimethylformamide dimethyl acetal (DMF-DMA, 1.5 mL, 11.2 mmol) and heated to 110° C. The reaction was run for 30 minutes and then dried, at which point acetic acid (0.12 mL) and ethanol (0.6 mL) were added, providing a clear solution. To this solution was added hydrazine hydrate (0.024 mL, 0.76 mmol) and the reaction was stirred for 30 minutes. The solution was filtered and purified using preparative HPLC to provide 172 (2.5 mg, 7.5% yield).
  • DMF-DMA N,N-dimethylformamide dimethyl acetal
  • Example 173 To slurry of Example 173 (50 mg, 0.085 mmol) and potassium carbonate (47.0 mg, 0.340 mmol) in DMF (0.3 mL) at room temperature was added iodoethane (19.90 mg, 0.128 mmol) and the resulting mixture was allowed to stir at room temperature for 3 h. A mixture of two regioisomers was seen; however, these were typically separable by preparative HPLC (exceptions noted in the table). Structural assignment was made by analysis of 1 H NMR compared to compounds with known (by synthesis or crystal structure) regiochemistry. The crude reaction mixture was diluted with DMSO and was subjected to purification by reverse-phase HPLC to afford fractions containing the major product.
  • Example 173 The following Examples were prepared using similar conditions as described for the preparation of Example 173 and Example 174:
  • Example 181 The following Examples were prepared using similar conditions as described for the preparation of Example 181 and Example 182:
  • Example 185 To slurry of the substrate Example 185 (25 mg, 0.061 mmol) and 1-bromo-2-fluoroethane (15.47 mg, 0.122 mmol) in DMF (0.3 mL) at room temperature was added 1-bromo-2-fluoroethane (15.47 mg, 0.122 mmol) stirred at room temperature for 3 h and 60° C. for an additional 3 h.
  • the crude reaction mixture was diluted with DMSO and was subjected to reverse-phase HPLC to afford fractions containing the desired product which were concentrated under vacuum to afford 2.5 mg of Example 186.
  • Example 186 The following Examples were prepared in a similar manner to Example 186:
  • 6-Chloro-4-((3-ethynyl-2-methoxyphenyl)amino)-N-methylpyridazine-3-carboxamide (prepared in Preparation 7) (25 mg, 0.078 mmol) was combined with benzoic acid (2 mg, 0.016 mmol), L-Ascorbic acid sodium salt (2 mg, 0.0010 mmol) and copper(II) sulfate (2 mg, 0.013 mmol) in a small flask.
  • a solution of 2-azidopropane (6.65 mg, 0.078 mmol) in tert-butyl alcohol (0.5 mL) and water (0.5 mL) was subsequently added and the reaction was stirred at room temperature for 1 hour.
  • the reaction was diluted with ethyl acetate, washed with water, saturated aqueous ammonium chloride and brine, and then dried over sodium sulfate, filtered and concentrated.
  • the crude product was re-dissolved in DMF and purified by preparative HPLC to provide 190 (15.4 mg, 57%).
  • Example 191 The following Examples were prepared in a similar manner to Example 191:
  • Example 192 was prepared in a similar manner to Example 191.
  • the resulting reddish-orange oil was then dissolved in ethanol (4 mL) and AcOH (4 mL) and cooled in an ice bath before adding hydrazine (as a monohydrate) (0.482 mL, 7.69 mmol). Let warm to room temperature then resulting solution was heated to 80° C. for 30 minutes before cooling and concentrating on the rotovap. The resulting material was diluted with water ( ⁇ 25 mL) which caused an oil to form from the solution. The mixture was cooled in an ice bath, sonicated, and then stirred vigorously which eventually cause the oil to solidify.
  • Example 195 35 mg, 0.085 mmol and cesium carbonate (83 mg, 0.256 mmol) were mixed in DMF (0.3 mL) and 2,2-dimethyloxirane (12.30 mg, 0.171 mmol) was added followed by heating the resulting mixture at 60° C. for overnight ( ⁇ 16 h). The mixture was cooled, dissolved in DMSO, filtered and was purified via preparative HPLC. Unless noted (table below) the major and minor regioisomers (assignment from unambiguous parallel synthesis of representative examples) were isolated and characterized separately containing the major product were combined and dried via centrifugal evaporation to afford 30.2 mg of Example 196.
  • Example 196 The following Examples were prepared in a similar manner to Example 196:
  • Example 200 was prepared in a similar manner to Example 195 by using 1,1-dimethoxy-N,N-dimethylethanamine in place of 1,1-dimethoxy-N,N-dimethylmethanamine in Step 3. Afforded Example 200 as a tan solid.
  • Int1 (1.14 g, 7.3 mmol) was placed in a 500 mL RBF and triethylamine (1.02 mL, 7.3 mmol) was added, followed by phosphorus oxychloride (9 mL, 97 mmol).
  • the flask was placed in a room temperature oil bath and once self-reflux ceased, the temperature was raised to 80° C. Once that temperature was reached and the vigorous reflux subsided the temperature was raised again to 110° C. and the reaction run for 120 minutes. The heating was stopped and the reaction allowed to cool to ⁇ 90° C.
  • the crude material was suspended in hot dichloromethane and absorbed onto CELITE®, the CELITE® was dried and the material was purified by automated chromatography. Following chromatography the collected product was suspended in hot dichloromethane, cooled and then filtered, rinsing with dichloromethane and then methanol, collecting the residual powder provided 201 (10 mg, 12% yield).
  • the mixture was treated with Pd 2 (dba) 3 (33 mg, 0.036 mmol) and the vessel was sealed and subjected to five evacuate-fill cycles with argon. The mixture was stirred on a heating block at 80° C. for 16 h. The mixture was cooled to room temperature and partitioned between water and ethyl acetate. The aqueous phase was extracted with ethyl acetate, and the combined organic phases were washed with brine, dried over sodium sulfate and concentrated under vacuum.
US16/201,653 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses Active USRE47929E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/201,653 USRE47929E1 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261723840P 2012-11-08 2012-11-08
US16/201,653 USRE47929E1 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses
PCT/US2013/068846 WO2014074661A1 (en) 2012-11-08 2013-11-07 AMIDE-SUBSTITUTED HETEROCYCLIC COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFN ALPHα RESPONSES
US14/441,183 US9505748B2 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses

Publications (1)

Publication Number Publication Date
USRE47929E1 true USRE47929E1 (en) 2020-04-07

Family

ID=49876960

Family Applications (6)

Application Number Title Priority Date Filing Date
US14/441,183 Ceased US9505748B2 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses
US16/201,653 Active USRE47929E1 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses
US15/289,437 Active US10000480B2 (en) 2012-11-08 2016-10-10 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US15/979,770 Active US10526321B2 (en) 2012-11-08 2018-05-15 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US16/687,279 Active US11021475B2 (en) 2012-11-08 2019-11-18 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US17/244,938 Pending US20220356180A1 (en) 2012-11-08 2021-04-29 Amide-substituted heterocyclic compounds useful as modulators of il-12, il-23 and/or ifn alpha responses

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/441,183 Ceased US9505748B2 (en) 2012-11-08 2013-11-07 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFNα responses

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/289,437 Active US10000480B2 (en) 2012-11-08 2016-10-10 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US15/979,770 Active US10526321B2 (en) 2012-11-08 2018-05-15 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US16/687,279 Active US11021475B2 (en) 2012-11-08 2019-11-18 Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US17/244,938 Pending US20220356180A1 (en) 2012-11-08 2021-04-29 Amide-substituted heterocyclic compounds useful as modulators of il-12, il-23 and/or ifn alpha responses

Country Status (38)

Country Link
US (6) US9505748B2 (pl)
EP (3) EP4071144A1 (pl)
JP (3) JP6407159B2 (pl)
KR (1) KR102195194B1 (pl)
CN (1) CN104884454B (pl)
AR (1) AR094452A1 (pl)
AU (4) AU2013341186B2 (pl)
BR (1) BR112015010102B1 (pl)
CA (1) CA2890981C (pl)
CL (1) CL2015001231A1 (pl)
CY (1) CY1121188T1 (pl)
DK (2) DK3495358T3 (pl)
EA (1) EA028814B1 (pl)
ES (2) ES2702148T3 (pl)
FI (1) FIC20230028I1 (pl)
FR (1) FR23C1030I1 (pl)
HK (1) HK1215255A1 (pl)
HR (2) HRP20220766T1 (pl)
HU (3) HUE059409T2 (pl)
IL (1) IL238610A0 (pl)
LT (3) LT2922846T (pl)
MA (1) MA38072A1 (pl)
MX (2) MX2015005731A (pl)
MY (2) MY175448A (pl)
NL (1) NL301238I2 (pl)
NZ (1) NZ708859A (pl)
PE (1) PE20150944A1 (pl)
PH (1) PH12015501004A1 (pl)
PL (2) PL3495358T3 (pl)
PT (2) PT2922846T (pl)
RS (2) RS63328B1 (pl)
SG (3) SG10201706985UA (pl)
SI (2) SI2922846T1 (pl)
TR (1) TR201820824T4 (pl)
TW (1) TWI605041B (pl)
UY (1) UY35126A (pl)
WO (1) WO2014074661A1 (pl)
ZA (1) ZA201504052B (pl)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11021475B2 (en) * 2012-11-08 2021-06-01 Bristol-Myers Squibb Company Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US11357775B2 (en) 2019-04-30 2022-06-14 Celgene Corporation Combination therapies comprising apremilast and Tyk2 inhibitors
WO2022165141A1 (en) 2021-01-29 2022-08-04 Bristol-Myers Squibb Company Crystal form of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
WO2022212181A1 (en) 2021-03-29 2022-10-06 Bristol-Myers Squibb Company Crystal forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
WO2022241172A1 (en) 2021-05-14 2022-11-17 Bristol-Myers Squibb Company Substituted heterocyclic compounds
WO2023023322A1 (en) 2021-08-20 2023-02-23 Bristol-Myers Squibb Company Crystal forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
US11613548B2 (en) 2021-02-19 2023-03-28 Sudo Biosciences Limited Substituted pyridines, pyridazines, pyrimidines, and 1,2,4-triazines as TYK2 inhibitors
WO2023049241A1 (en) 2021-09-23 2023-03-30 Bristol-Myers Squibb Company Methods of treating hair-loss disorders with tyk2 inhibitors
WO2023055901A2 (en) 2021-09-30 2023-04-06 Bristol-Myers Squibb Company Methods for determining responsiveness to tyk2 inhibitors
WO2023076515A1 (en) 2021-10-28 2023-05-04 Bristol-Myers Squibb Company Topical formulations of deucravacitinib

Families Citing this family (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2917186B1 (en) 2012-11-08 2017-10-18 Bristol-Myers Squibb Company Alkyl amide-substituted pyrimidine compounds useful in the modulation of il-12, il-23 and/or ifn
AR094537A1 (es) 2013-11-07 2015-08-12 Bristol Myers Squibb Co COMPUESTOS DE PIRIDILO SUSTITUIDOS CON ALQUILAMIDA ÚTILES COMO MODULADORES DE LAS RESPUESTAS DE IL-12, IL-23 Y/O IFNa
US10273237B2 (en) 2013-12-10 2019-04-30 Bristol-Myers Squibb Company Imidazopyridazine compounds useful as modulators of IL-12, IL-23 and/or IFN-α responses
WO2015123453A1 (en) * 2014-02-14 2015-08-20 Portola Pharmaceuticals, Inc. Pyridazine compounds as jak inhibitors
ES2916582T3 (es) 2015-11-26 2022-07-01 Novartis Ag Derivados de diaminopiridina
EP3848370A3 (en) 2016-10-14 2021-09-15 Nimbus Lakshmi, Inc. Tyk2 inhibitors and uses thereof
CN110300590A (zh) 2016-10-21 2019-10-01 林伯士拉克许米公司 Tyk2抑制剂及其用途
EP3555111B1 (en) 2016-12-13 2022-01-26 Bristol-Myers Squibb Company Phosphine oxide alkyl amide substituted heteroaryl compounds as modulators of il-12, il-23, and/or ifn alpha responses
JOP20180011A1 (ar) 2017-02-16 2019-01-30 Gilead Sciences Inc مشتقات بيرولو [1، 2-b]بيريدازين
TW202321243A (zh) 2017-03-08 2023-06-01 美商林伯士拉克許米公司 Tyk2抑制劑之生產方法
EP3601268B1 (en) * 2017-03-30 2021-03-17 Bristol-Myers Squibb Company Process for the preparation of 6-(cyclopropaneamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
PE20211461A1 (es) * 2017-11-21 2021-08-05 Bristol Myers Squibb Co Compuestos de heteroarilo sustituidos con alquilamida de piridinsulfona
US10508113B2 (en) 2018-03-12 2019-12-17 Abbvie Inc. Inhibitors of tyrosine kinase 2 mediated signaling
CN111936486B (zh) * 2018-03-22 2023-09-22 百时美施贵宝公司 用作IL-12、IL-23和/或IFNα响应的调节剂的包含吡啶的杂环化合物
CN112236425A (zh) * 2018-05-31 2021-01-15 百时美施贵宝公司 6-(环丙烷甲酰氨基)-4-((2-甲氧基-3-(1-甲基-1h-1,2,4-***-3-基)苯基)氨基)-n-(甲基-d3)哒嗪-3-甲酰胺的结晶形式
TW202136268A (zh) 2018-07-13 2021-10-01 美商基利科學股份有限公司 吡咯并[1,2-b]嗒𠯤衍生物
GB201816369D0 (en) 2018-10-08 2018-11-28 Sareum Ltd Pharmaceutical compounds
AU2019360941A1 (en) 2018-10-15 2021-04-29 Takeda Pharmaceutical Company Limited TYK2 inhibitors and uses thereof
EP3870579A4 (en) * 2018-10-22 2022-10-19 Alumis Inc. TYK2 INHIBITORS AND THEIR USES
BR112021007861A2 (pt) * 2018-10-30 2021-08-03 Bristol-Myers Squibb Pharma Company compostos heterocíclicos substituídos por amida
US11053241B2 (en) 2018-11-30 2021-07-06 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
EP3914357A4 (en) 2019-01-23 2022-10-12 Nimbus Lakshmi, Inc. TYK2 INHIBITORS AND THEIR USES
CN111757878B (zh) * 2019-01-28 2023-07-28 江苏豪森药业集团有限公司 一种哒嗪类衍生物抑制剂、其制备方法和应用
CN111484480B (zh) * 2019-01-29 2023-08-11 上海翰森生物医药科技有限公司 一种多环类衍生物抑制剂、其制备方法和应用
CA3128014A1 (en) * 2019-01-30 2020-08-06 Bristol-Myers Squibb Company Amide-disubstituted pyridine or pyridazine compounds
SG11202112043PA (en) * 2019-04-30 2021-11-29 Celgene Corp Combination therapies comprising apremilast and tyk2 inhibitors
EP3983086A1 (en) * 2019-06-12 2022-04-20 Bristol-Myers Squibb Company Crystalline salt forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3) pyridazine-3-carboxamide
US20230167082A1 (en) 2019-07-16 2023-06-01 Bristol-Myers Squibb Company Prodrugs in the modulation of interleukin
JP2022542704A (ja) * 2019-08-01 2022-10-06 インテグラル バイオサイエンシーズ プライベート リミテッド キナーゼインヒビターとしての複素環式化合物およびその使用
KR20220105631A (ko) 2019-09-13 2022-07-27 님버스 새턴 인코포레이티드 Hpk1 길항제 및 이의 용도
KR20220066105A (ko) * 2019-09-18 2022-05-23 브리스톨-마이어스 스큅 컴퍼니 Tyk2 억제제를 위한 연장 방출 투여 형태
CN114787152A (zh) * 2019-12-27 2022-07-22 苏州科睿思制药有限公司 一种bms-986165晶型及其制备方法和用途
CN114787154A (zh) * 2020-01-19 2022-07-22 苏州科睿思制药有限公司 一种Deucravacitinib的晶型及其制备方法和用途
WO2021143430A1 (zh) * 2020-01-19 2021-07-22 苏州科睿思制药有限公司 一种bms-986165盐酸盐晶型及其制备方法和用途
WO2021170046A1 (en) * 2020-02-26 2021-09-02 Beigene, Ltd. Tyk-2 inhibitor
AU2021234496A1 (en) * 2020-03-11 2022-10-20 Beijing Innocare Pharma Tech Co., Ltd. Heterocyclic compounds for inhibiting TYK2 activities
WO2021204626A1 (en) * 2020-04-06 2021-10-14 Almirall, S.A. Aryl and heteroaryl-carboxamide substituted heteroaryl compounds as tyk2 inhibitors
GB202005114D0 (en) 2020-04-07 2020-05-20 Sareum Ltd Crystalline Forms of a Pharmaceutical Compound
US20230167092A1 (en) * 2020-04-28 2023-06-01 Bristol-Myers Squibb Company Substituted n-(methyl-d3)pyridazine-3-carboxamide or n-(methyl-d3)-nicotinamide compounds as il-12, il-23 and/or ifnalpha modulators
CN113563309A (zh) * 2020-04-28 2021-10-29 浙江海正药业股份有限公司 吡啶类衍生物及其制备方法和用途
CN113735837B (zh) * 2020-05-28 2023-09-01 江苏先声药业有限公司 哒嗪类化合物及其用途
CN113735836B (zh) * 2020-05-28 2023-05-30 江苏先声药业有限公司 哒嗪类化合物及其应用
CN113773262A (zh) * 2020-06-09 2021-12-10 江苏先声药业有限公司 哒嗪类化合物
CA3186000A1 (en) * 2020-07-24 2022-01-27 Shanghai Hansoh Biomedical Co., Ltd. Crystal form of pyridazine derivative free base, and preparation method therefor and use thereof
EP3944859A1 (en) 2020-07-30 2022-02-02 Assistance Publique Hôpitaux de Paris Method for treating immune toxicities induced by immune checkpoint inhibitors
WO2022021684A1 (zh) * 2020-07-31 2022-02-03 苏州科睿思制药有限公司 一种bms-986165盐酸盐晶型csv及其制备方法和用途
CN116472044A (zh) 2020-09-18 2023-07-21 百时美施贵宝公司 包含可溶胀核芯的Tyk2抑制剂的剂型
CN116615200A (zh) * 2020-10-20 2023-08-18 杭州领业医药科技有限公司 哒嗪衍生物的晶型
WO2022100710A1 (en) * 2020-11-12 2022-05-19 Cullgen (Shanghai) , Inc. Tyrosine kinase 2 (tyk2) degradation compounds and methods of use
TW202227430A (zh) * 2020-11-17 2022-07-16 大陸商江蘇恆瑞醫藥股份有限公司 含氮雜環類衍生物、其製備方法及其在醫藥上的應用
WO2022121868A1 (zh) * 2020-12-08 2022-06-16 正大天晴药业集团股份有限公司 含酰胺基和杂环烷基的tyk2抑制剂化合物
AU2022216810A1 (en) 2021-02-02 2023-08-24 Liminal Biosciences Limited Gpr84 antagonists and uses thereof
AU2022215844A1 (en) 2021-02-02 2023-09-14 Liminal Biosciences Limited Gpr84 antagonists and uses thereof
TW202233600A (zh) * 2021-02-06 2022-09-01 大陸商正大天晴藥業集團股份有限公司 含聯環的tyk2抑制劑化合物、藥物組合物及其用途
JP2024509192A (ja) 2021-03-05 2024-02-29 ニンバス サターン, インコーポレイテッド Hpk1アンタゴニスト及びその使用
WO2022193499A1 (en) * 2021-03-16 2022-09-22 Anrui Biomedical Technology (Guangzhou) Co., Ltd. Amino heteroaryl compounds and compositions
CN115141149A (zh) * 2021-03-30 2022-10-04 浙江文达医药科技有限公司 作为tyk2假激酶结构域抑制剂的杂环化合物及合成方法和用途
CN115197196A (zh) * 2021-04-06 2022-10-18 扬子江药业集团有限公司 Tyk2抑制剂及其用途
AU2022268464A1 (en) 2021-05-04 2023-12-21 Shanghai Zheye Biotechnology Co. Ltd. Nitrogen-containing heterocyclic pyridine compound
TW202311247A (zh) * 2021-05-14 2023-03-16 美商必治妥美雅史谷比公司 經取代之雜環化合物
EP4337650A1 (en) 2021-05-14 2024-03-20 Bristol-Myers Squibb Company Substituted heterocyclic compounds
KR20240007242A (ko) * 2021-05-14 2024-01-16 브리스톨-마이어스 스큅 컴퍼니 치환된 헤테로시클릭 화합물
WO2022253335A1 (zh) * 2021-06-02 2022-12-08 南京明德新药研发有限公司 含磺酰基的芳基类化合物及其应用
BR112023026964A2 (pt) * 2021-06-22 2024-03-12 Medshine Discovery Inc Composto de sulfoximina e uso do mesmo
WO2023284869A1 (zh) * 2021-07-15 2023-01-19 南京明德新药研发有限公司 含硫/磷的芳基类化合物及其应用
WO2023027948A1 (en) * 2021-08-21 2023-03-02 Relay Therapeutics, Inc. Jak2 inhibitors and methods of use thereof
WO2023076161A1 (en) 2021-10-25 2023-05-04 Kymera Therapeutics, Inc. Tyk2 degraders and uses thereof
WO2023102085A1 (en) 2021-12-01 2023-06-08 Teva Czech Industries S.R.O. Solid state forms of deucravacitinib, deucravacitinib hcl and process for preparation of deucravacitinib and intermediates
WO2023108536A1 (en) * 2021-12-16 2023-06-22 Lynk Pharmaceuticals Co. Ltd. Tyk2 inhibitors and compositions and methods thereof
WO2023109954A1 (en) * 2021-12-16 2023-06-22 Lynk Pharmaceuticals Co. Ltd. Tyk2 inhibitors and compositions and methods thereof
EP4206196A1 (en) 2021-12-29 2023-07-05 Almirall S.A. Pyrimidine substituted derivatives as tyk2 inhibitors
CN116693449A (zh) * 2022-03-04 2023-09-05 上海致根医药科技有限公司 用作tyk2抑制剂的化合物、其制备方法及其在医药上的应用
WO2023231997A1 (zh) * 2022-05-31 2023-12-07 上海华汇拓医药科技有限公司 一种哒嗪类化合物、其制备方法和用途
WO2024020221A1 (en) 2022-07-21 2024-01-25 Arvinas Operations, Inc. Modulators of tyk2 proteolysis and associated methods of use
WO2024028365A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Substituted pyridone gpr84 antagonists and uses thereof
WO2024028363A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Heteroaryl carboxamide and related gpr84 antagonists and uses thereof
WO2024028364A1 (en) 2022-08-02 2024-02-08 Liminal Biosciences Limited Aryl-triazolyl and related gpr84 antagonists and uses thereof
CN116284040A (zh) * 2023-01-05 2023-06-23 华润医药研究院(深圳)有限公司 含氮杂环类化合物及其医药用途
CN116162093B (zh) * 2023-04-25 2023-06-23 中南大学湘雅医院 一种tyk2抑制剂化合物及其用途

Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075113A1 (fr) 1999-06-09 2000-12-14 Yamanouchi Pharmaceutical Co., Ltd. Nouveaux derives carboxamide heterocycliques
WO2000076980A1 (fr) 1999-06-10 2000-12-21 Yamanouchi Pharmaceutical Co., Ltd. Nouveaux derives heterocycliques azotes ou leurs sels
US20020061865A1 (en) 2000-10-03 2002-05-23 Kramer James Bernard Pyridotriazines and pyridopyridazines
WO2004007682A2 (en) 2002-07-15 2004-01-22 Wyeth Methods and compositions for modulating t helper (th) cell development and function
US20040142930A1 (en) 2000-04-28 2004-07-22 Koichiro Yamada Cyclic compounds
WO2004106293A2 (en) 2003-05-21 2004-12-09 Bristol-Myers Squibb Company Oxazolyl - and thiazolyl - purine based tricyclic compounds.
WO2011113802A2 (en) 2010-03-17 2011-09-22 F. Hoffmann-La Roche Ag Imidazopyridine compounds, compositions and methods of use
WO2012035039A1 (en) 2010-09-15 2012-03-22 F. Hoffmann-La Roche Ag Azabenzothiazole compounds, compositions and methods of use
WO2012066061A1 (en) 2010-11-19 2012-05-24 F. Hoffmann-La Roche Ag Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
WO2013047813A1 (ja) 2011-09-30 2013-04-04 大鵬薬品工業株式会社 1,2,4-トリアジン-6-カルボキサミド誘導体
WO2013104573A1 (en) 2012-01-10 2013-07-18 F. Hoffmann-La Roche Ag Pyridazine amide compounds and their use as syk inhibitors
WO2013171690A1 (en) 2012-05-16 2013-11-21 Novartis Ag Monocyclic heteroaryl cycloalkyldiamine derivatives
WO2013192049A2 (en) 2012-06-22 2013-12-27 Portola Pharmaceuticals, Inc. 1,2,4-triazine-6-carboxamide kinase inhibitors
WO2014060371A1 (en) 2012-10-19 2014-04-24 F. Hoffmann-La Roche Ag Inhibitors of syk
WO2014074660A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company ALKYL-AMIDE-SUBSTITUTED PYRIDYL COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFNα RESPONSES
WO2014074661A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company AMIDE-SUBSTITUTED HETEROCYCLIC COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFN ALPHα RESPONSES
WO2014074670A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company ALKYL AMIDE-SUBSTITUTED PYRIMIDINE COMPOUNDS USEFUL IN THE MODULATION OF IL-12, IL-23 AND/OR IFNα
WO2015069310A1 (en) 2013-11-07 2015-05-14 Bristol-Myers Squibb Company Alkyl-amide-substituted pyridyl compounds useful as modulators of il-12, il-23 and/or ifnalpha responses
WO2018183649A1 (en) 2017-03-30 2018-10-04 Bristol-Myers Squibb Company Process for the preparation of 6-(cyclopropaneamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4622047B2 (ja) * 1999-06-09 2011-02-02 アステラス製薬株式会社 新規なヘテロ環カルボキサミド誘導体

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075113A1 (fr) 1999-06-09 2000-12-14 Yamanouchi Pharmaceutical Co., Ltd. Nouveaux derives carboxamide heterocycliques
EP1184376A1 (en) 1999-06-09 2002-03-06 Yamanouchi Pharmaceutical Co. Ltd. Novel heterocyclic carboxamide derivatives
WO2000076980A1 (fr) 1999-06-10 2000-12-21 Yamanouchi Pharmaceutical Co., Ltd. Nouveaux derives heterocycliques azotes ou leurs sels
US20040142930A1 (en) 2000-04-28 2004-07-22 Koichiro Yamada Cyclic compounds
US20020061865A1 (en) 2000-10-03 2002-05-23 Kramer James Bernard Pyridotriazines and pyridopyridazines
WO2004007682A2 (en) 2002-07-15 2004-01-22 Wyeth Methods and compositions for modulating t helper (th) cell development and function
WO2004106293A2 (en) 2003-05-21 2004-12-09 Bristol-Myers Squibb Company Oxazolyl - and thiazolyl - purine based tricyclic compounds.
WO2011113802A2 (en) 2010-03-17 2011-09-22 F. Hoffmann-La Roche Ag Imidazopyridine compounds, compositions and methods of use
WO2012035039A1 (en) 2010-09-15 2012-03-22 F. Hoffmann-La Roche Ag Azabenzothiazole compounds, compositions and methods of use
WO2012066061A1 (en) 2010-11-19 2012-05-24 F. Hoffmann-La Roche Ag Pyrazolopyridines and pyrazolopyridines and their use as tyk2 inhibitors
WO2013047813A1 (ja) 2011-09-30 2013-04-04 大鵬薬品工業株式会社 1,2,4-トリアジン-6-カルボキサミド誘導体
WO2013104573A1 (en) 2012-01-10 2013-07-18 F. Hoffmann-La Roche Ag Pyridazine amide compounds and their use as syk inhibitors
WO2013171690A1 (en) 2012-05-16 2013-11-21 Novartis Ag Monocyclic heteroaryl cycloalkyldiamine derivatives
WO2013192049A2 (en) 2012-06-22 2013-12-27 Portola Pharmaceuticals, Inc. 1,2,4-triazine-6-carboxamide kinase inhibitors
WO2014060371A1 (en) 2012-10-19 2014-04-24 F. Hoffmann-La Roche Ag Inhibitors of syk
WO2014074660A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company ALKYL-AMIDE-SUBSTITUTED PYRIDYL COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFNα RESPONSES
WO2014074661A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company AMIDE-SUBSTITUTED HETEROCYCLIC COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFN ALPHα RESPONSES
WO2014074670A1 (en) 2012-11-08 2014-05-15 Bristol-Myers Squibb Company ALKYL AMIDE-SUBSTITUTED PYRIMIDINE COMPOUNDS USEFUL IN THE MODULATION OF IL-12, IL-23 AND/OR IFNα
WO2015069310A1 (en) 2013-11-07 2015-05-14 Bristol-Myers Squibb Company Alkyl-amide-substituted pyridyl compounds useful as modulators of il-12, il-23 and/or ifnalpha responses
WO2018183649A1 (en) 2017-03-30 2018-10-04 Bristol-Myers Squibb Company Process for the preparation of 6-(cyclopropaneamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide

Non-Patent Citations (55)

* Cited by examiner, † Cited by third party
Title
Barder, T. E. et al., "Catalysts for Suzuki-Miyaura Coupling Processes: Scope and Studies of the Effect of Ligand Structure," Journal of the American Chemical Society, 127:4685-4696 (2005).
Bave, U. et al., "Activation of the Type I Interferon System in Primary Sjogren's Syndrome: A Possible Etiopathogenic Mechanism," Arthritis Rheum., 52:1185-1195 (2005).
Bengtsson, A. A. et al., "Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies," Lupus, 9:664-671 (2000).
Bennett, L. et al., "Interferon and Granulopoiesis Signatures in Systemic Lupus Erythematosus Blood," The Journal of Experimental Medicine, 197:711-723 (2003).
Bundgaard, H., "Design and Application of Prodrugs," Krosgaard-Larsen, P. et al eds., A Textbook of Drug Design and Development, 113-191, Harwood Academic Publishers (1991).
Bundgaard, H., "Penetration Enhancement for Polypeptides through Epithelia: (C) Means to Enhance Penetration," Advanced Drug Delivery Reviews, 8:1-38 (1992).
Bundgaard, H., ed., Design of Prodrugs, Elsevier (1985).
Caron, S., "Synthesis of Common Aromatic Heterocycles," Practical Synthetic Organic Chemistry, 609-647 (2011).
Chinchilla, R. et al., "Recent advances in Sonogashira reactions," The Royal Society of Chemistry, 40:5084-5121 (2011).
Chinchilla, R. et al., "The Sonogashira Reaction: A Booming Methodology in Synthetic Organic Chemistry," Chemical Reviews 107:874-923 (2007).
Cho, J. H. et al., "Recent insights into the genetics of inflammatory bowel disease," Gastroenterology, 140:1704-1712 (2011).
Couturier, N. et al., "Tyrosine kinase 2 variant influences T lymphocyte polarization and multiple sclerosis susceptibility," Brain, 134:693-703 (2011).
Cua, D. J. et al "Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain," Nature, 421:744-748 (2003).
Deng, Y. et al., "Genetic susceptibility to systemic lupus erythematosus in the genomic era," Nat. Rev. Rheumatol., 6:683-692 (2010).
Dyckman et al., "Imidazo[4,5-d]thiazolo[5,4-b]pyridine based inhibitors of IKK2: Synthesis, SAR, PK/PD and activity in a preclinical model of rheumatoid arthritis," Bioorganic and Medicinal Chemistry Letters, 383-386 (2011).
El-Ahl, A-A. S. et al., "A novel approach for the synthesis of 5-substituted tetrazole derivatives from primary amides in mild one-step method," Tetrahedron Letters, 38:1257-1260 (1997).
Ellinghaus, D. et al., "Combined Analysis of Genome-wide Association Studies for Crohn Disease and Psoriasis Identifies Seven Shared Susceptibility Loci," The American Society of Human Genetics, 90:636-647 (2012).
Extended European Search Report dated Feb. 22, 2019, issued in Application No. 18197550.9-1116.
Eyre, S. et al., "High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis," Nat. Genet., 44:1336-1340 (2012).
Garnier, E. et al., "New Polynitrogen Materials Based on Fused 1,2,4-Triazines," Synlett, 472-474 (2006).
Gottlieb, A. et al., "Ustekinumab, a human interleukin 12/23 monoclonal antibody, for psoriatic arthritis: randomized, double-blind, placebo-controlled, crossover trial," Lancet, 373:633-640 (2009).
Gracie, J. A. et al., "Interleukin-12 induces interferon-gamma-dependent switching of IgG alloantibody subclass," Eur. J. Immunol., 26:1217-1221 (1996).
Graham, D. et al., "Association of polymorphisms across the tyrosine kinase gene, TYK2 in UK SLE families," Rheumatology (Oxford), 46:927-930 (2007).
Grecian, S. et al., "Ruthenium-Catalyzed Cycloaddition of Nitrile Oxides and Alkynes: Practical Synthesis of lsoxazoles," Angew. Chem. Int. Ed., 47:8285-8287 (2008).
Hall, J. C. et al., "Type I interferons: crucial participants in disease amplification in autoimmunity," Nat. Rev. Rheumatol., 6:40-49 (2010).
Hong, K. et al., "IL-12, Independently of IFN-γ, Plays a Crucial Role in the Pathogenesis of a Murine Psoriasis-Like Skin Disorder," The Journal of Immunology, 162:7480-7491 (1999).
Huang, X. et al., "Dysregulated expression of interleukin-23 and interleukin-12 subunits in systemic lupus erythematosus patients," Mod. Rheumatol., 17:220-223 (2007).
Hue, S. et al., "Interleukin-23 drives innate and T cell-mediated intestinal inflammation," J. Exp. Med., 203:2473-2483 (2006).
Ishiyama, T. et al., "Palladium(0)-Catalyzed Cross-Coupling Reaction of Alkoxydiboron with Haloarenes: A Direct Procedure for Arylboronic Esters," The Journal of Organic Chemistry, 60:7508 (1995).
Kim, D. et al., "Induction of Interferon-a by Scleroderma Sera Containing Autoantibodies to Topoisomerase I: Association of Higher Interferon-α Activity With Lung Fibrosis," Arthritis Rheum., 58:2163-2173 (2008).
Kolb, H. C. et al., "Click Chemistry: Diverse Chemical Function from a Few Good Reactions," Angew. Chem. Int. Ed., 40:2004-2021 (2001).
Kyttaris, V. C. et al., "Cutting Edge: IL-23 Receptor Deficiency Prevents the Development of Lupus Nephritis in C57BL/6-lpr/lpr mice," The Journal of Immunology, 184:4605-4609 (2010).
Lee, E. et al.,"Increased Expression of Interleukin 23 p19 and p40 in Lesional Skin of Patients with Psoriasis Vulgaris," The Journal of Experimental Medicine, 199:125-130 (2004).
Lees, C. W. et al., "New IBD genetics: common pathways with other diseases," Gut, 60:1739-1753 (2011).
Leonardi, C. L. et al., "Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, 40 placebo-controlled trial (Phoenix 1)," Lancet, 371:1665-1674 (2008).
lshizaki, M. et al., "Involvement of Tyrosine Kinase-2 in Both the IL-12/Th1 and IL-23/Th17 Axes In Vivo," The Journal of Immunology, 187:181-189 (2011).
McGeachy, M. J. et al., "The link between IL-23 and Th17 cell-mediated immune pathologies," Semin. Immunol., 19:372-376 (2007).
Meldal, M. et al., "Cu-Catalyzed Azide-Alkyne Cycloaddition" American Chemical Society, 108:2952-3015 (2008).
Minegishi, Y. et al., "Human Tyrosine Kinase 2 Deficiency Reveals its Requisite Roles in Multiple Cytokine Signals Involved in Innate and Acquired Immunity," Immunity, 25:745-755 (2006).
Miyaura, N. et al., "Palladium-Catalyzed Cross-Coupling Reactions of Organoboron Compounds," American Chemical Society, 95:2457-2483 (1995).
Monnier, F. et al., "Catalytic C-C, C-N, and C-O Ullmann-Type Coupling Reactions," Angew. Chem. Int. Ed., 48:6954-6971 (2009).
Murphy, C. A. et al., "Divergent Pro- and Anti-Inflammatory Roles for IL-23 and IL-12 in Joint Autoimmune Inflammation," The Journal of Experimental Medicine, 198:1951-1957 (2003).
Oyamada, A. et al., "Tyrosine Kinase 2 Plays Critical Roles in the Pathogenic CD4 T Cell Responses for the Development of Experimental Autoimmune Encephalomyelitis," The Journal of Immunology, 183:7539-7546 (2009).
Peterson, K. S. et al., "Characterization of Heterogeneity in the Molecular Pathogenesis of Lupus Nephritis from Transcriptional Profiles of Laser-captured Glomeruli," The Journal of Clinical Investigation, 113:1722-1733 (2004).
Prchal-Murphy, M. et al., "TYK2 Kinase Activity is Required for Functional Type I Interferon Responses In Vivo," PLoS One, 7:e39141 (2012).
Sandborn, W. J. et al., "Ustekinumab Crohn's Disease Study Group. A randomized trial of Ustekinumab, A Human Interleukin-12/23 Monoclonal Antibody, in Patients with Moderate-to-Severe Crohn's Disease," Gastroenterology, 135:1130-1141 (2008).
Sandling, J. K. et al., "A Candidate Gene Study of the Type I Interferon Pathway Implicates IKBKE and IL8 as Risk Loci for SLE," European Journal of Human Genetics, 19:479-484 (2011).
Santiago-Raber, M. L. et al., "Type-I interferon Receptor Deficiency Reduces Lupus-Like Disease in NZB Mice," J. Exp. Med., 197:777-788 (2003).
Schroder, K. et al., "Interferon-γ: an overview of signals, mechanisms and functions," J. Leukoc. Biol., 75(2):163-189 (2004).
Stanforth, S. P., "Catalytic Cross-coupling Reactions in Biaryl Synthesis," Tetrahedron, 54:263- 303 (1998).
Surry, D.S. et al., "Dialkylbiaryl phosphines in Pd-catalyzed amination: a user's guide," Chemical Science, 2:27-50 (2011).
Tao, J. H. et al., "Meta-analysis of TYK2 gene polymorphisms association with susceptibility to autoimmune and inflammatory diseases," Mol. Biol. Rep., 38:4663-4672 (2011).
Tucci, M. et al., "Overexpression of interleukin-12 and T helper 1 predominance in lupus nephritis," Clinical Experimental Immunology, 154:247-254 (2008).
Tzartos, J. S. et al., "Interleukin-17 Production in Central Nervous System Infiltrating T Cells and Glial Cells is Associated with Active Disease in Multiple Sclerosis," The American Journal of Pathology, 172:146-155 (2008).
Widder, K. et al., eds., Methods in Enzymology, 112:309-396, Academic Press (1985).

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11021475B2 (en) * 2012-11-08 2021-06-01 Bristol-Myers Squibb Company Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US11357775B2 (en) 2019-04-30 2022-06-14 Celgene Corporation Combination therapies comprising apremilast and Tyk2 inhibitors
WO2022165141A1 (en) 2021-01-29 2022-08-04 Bristol-Myers Squibb Company Crystal form of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
US11613548B2 (en) 2021-02-19 2023-03-28 Sudo Biosciences Limited Substituted pyridines, pyridazines, pyrimidines, and 1,2,4-triazines as TYK2 inhibitors
WO2022212181A1 (en) 2021-03-29 2022-10-06 Bristol-Myers Squibb Company Crystal forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
WO2022241172A1 (en) 2021-05-14 2022-11-17 Bristol-Myers Squibb Company Substituted heterocyclic compounds
WO2023023322A1 (en) 2021-08-20 2023-02-23 Bristol-Myers Squibb Company Crystal forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyl-d3)pyridazine-3-carboxamide
WO2023049241A1 (en) 2021-09-23 2023-03-30 Bristol-Myers Squibb Company Methods of treating hair-loss disorders with tyk2 inhibitors
WO2023055901A2 (en) 2021-09-30 2023-04-06 Bristol-Myers Squibb Company Methods for determining responsiveness to tyk2 inhibitors
WO2023076515A1 (en) 2021-10-28 2023-05-04 Bristol-Myers Squibb Company Topical formulations of deucravacitinib

Also Published As

Publication number Publication date
JP2020002157A (ja) 2020-01-09
CN104884454A (zh) 2015-09-02
MY194668A (en) 2022-12-12
MA38072A1 (fr) 2016-07-29
PL2922846T3 (pl) 2019-03-29
US9505748B2 (en) 2016-11-29
BR112015010102B1 (pt) 2022-02-08
RS58187B1 (sr) 2019-03-29
US20150299183A1 (en) 2015-10-22
HUE041750T2 (hu) 2019-05-28
ES2914793T3 (es) 2022-06-16
US20180265504A1 (en) 2018-09-20
FIC20230028I1 (fi) 2023-08-28
AU2013341186B2 (en) 2017-03-30
EP2922846A1 (en) 2015-09-30
NZ708859A (en) 2018-11-30
TW201422593A (zh) 2014-06-16
RS63328B1 (sr) 2022-07-29
TWI605041B (zh) 2017-11-11
ZA201504052B (en) 2017-11-29
HUE059409T2 (hu) 2022-11-28
US20200071315A1 (en) 2020-03-05
DK2922846T3 (en) 2019-01-21
EA201590917A1 (ru) 2015-09-30
WO2014074661A1 (en) 2014-05-15
MX2015005731A (es) 2015-09-16
SI3495358T1 (sl) 2022-06-30
SG10201706897TA (en) 2017-09-28
CL2015001231A1 (es) 2015-08-28
MX2020003156A (es) 2020-07-29
MY175448A (en) 2020-06-29
JP6407159B2 (ja) 2018-10-17
SG11201503399XA (en) 2015-05-28
PT3495358T (pt) 2022-06-02
UY35126A (es) 2014-05-30
PT2922846T (pt) 2018-12-19
AU2013341186A1 (en) 2015-07-02
SG10201706985UA (en) 2017-10-30
HUS2300025I1 (hu) 2023-08-28
HRP20220766T1 (hr) 2022-09-16
TR201820824T4 (tr) 2019-01-21
US20220356180A1 (en) 2022-11-10
JP6585231B2 (ja) 2019-10-02
US20170022192A1 (en) 2017-01-26
EP3495358A1 (en) 2019-06-12
AR094452A1 (es) 2015-08-05
EP3495358B1 (en) 2022-04-27
HK1215255A1 (zh) 2016-08-19
LT2922846T (lt) 2018-12-10
CA2890981A1 (en) 2014-05-15
FR23C1030I1 (fr) 2023-10-13
DK3495358T3 (da) 2022-05-23
AU2018267545A1 (en) 2018-12-13
JP2016506369A (ja) 2016-03-03
AU2020203967B2 (en) 2020-10-22
EP2922846B1 (en) 2018-10-03
NL301238I2 (nl) 2023-10-04
EA028814B1 (ru) 2018-01-31
CN104884454B (zh) 2017-11-28
BR112015010102A2 (pt) 2017-07-11
AU2020203967A1 (en) 2020-07-16
PH12015501004B1 (en) 2015-07-27
US11021475B2 (en) 2021-06-01
EP3495358B8 (en) 2022-06-08
US10000480B2 (en) 2018-06-19
IL238610A0 (en) 2015-06-30
KR20150081339A (ko) 2015-07-13
CY1121188T1 (el) 2020-05-29
EP4071144A1 (en) 2022-10-12
PE20150944A1 (es) 2015-06-20
AU2018267545B2 (en) 2020-04-02
AU2017201076B2 (en) 2018-11-15
ES2702148T3 (es) 2019-02-27
AU2017201076A1 (en) 2017-03-09
JP2018154636A (ja) 2018-10-04
HRP20181937T1 (hr) 2019-01-25
LTPA2023523I1 (pl) 2023-09-11
PL3495358T3 (pl) 2022-06-27
SI2922846T1 (sl) 2019-04-30
US10526321B2 (en) 2020-01-07
KR102195194B1 (ko) 2020-12-24
PH12015501004A1 (en) 2015-07-27
LT3495358T (lt) 2022-05-25
CA2890981C (en) 2022-07-05

Similar Documents

Publication Publication Date Title
US11021475B2 (en) Amide-substituted heterocyclic compounds useful as modulators of IL-12, IL-23 and/or IFN alpha responses
US9987266B2 (en) Alkyl-amide-substituted pyridyl compounds useful as modulators of IL-12, IL-23 and/or IFNα responses
US9315494B2 (en) Alkyl-amide-substituted pyridyl compounds useful as modulators of IL-12, IL-23 and/or IFNα responses

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOSLIN, RYAN M.;WEINSTEIN, DAVID S.;WROBLESKI, STEPHEN T.;AND OTHERS;SIGNING DATES FROM 20131205 TO 20131217;REEL/FRAME:056025/0891

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIN, SHUQUN;SPERGEL, STEVEN H.;ZHANG, YANLEI;REEL/FRAME:056026/0027

Effective date: 20150219

CC Certificate of correction