WO2016007722A1 - Triazolopyridines and triazolopyrazines as lsd1 inhibitors - Google Patents

Triazolopyridines and triazolopyrazines as lsd1 inhibitors Download PDF

Info

Publication number
WO2016007722A1
WO2016007722A1 PCT/US2015/039706 US2015039706W WO2016007722A1 WO 2016007722 A1 WO2016007722 A1 WO 2016007722A1 US 2015039706 W US2015039706 W US 2015039706W WO 2016007722 A1 WO2016007722 A1 WO 2016007722A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
independently selected
haloalkyl
halo
optionally substituted
Prior art date
Application number
PCT/US2015/039706
Other languages
French (fr)
Inventor
Liangxing Wu
Xiaozhao Wang
Wenqing Yao
Colin Zhang
Original Assignee
Incyte Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Corporation filed Critical Incyte Corporation
Publication of WO2016007722A1 publication Critical patent/WO2016007722A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention is directed to [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5- a]pyrazine derivatives which are LSD1 inhibitors useful in the treatment of diseases such as cancer.
  • Epigenetic modifications can impact genetic variation but, when dysregulated, can also contribute to the development of various diseases (Portela, A. and M. Esteller, Epigenetic modifications and human disease. Nat Biotechnol, 2010. 28(10): p. 1057-68; Lund, A.H. and M. 15 van Lohuizen, Epigenetics and cancer. Genes Dev, 2004. 18(19): p. 2315-35). Recently, in depth cancer genomics studies have discovered many epigenetic regulatory genes are often mutated or their own expression is abnormal in a variety of cancers (Dawson, M.A. and T.
  • LSD1 lysine specific 25 demethylase-1
  • FAD flavin adenine dinucleotide
  • LSD1 lysine-specific demethylase 1
  • LSD2 Another lysine specific demethylase (LSD2) (Karytinos, A., et al., A novel mammalian flavin-dependent histone demethylase. J Biol Chem, 2009. 284(26): p. 17775-82).
  • Jumonji domain containing family that can demethylate mono-, di-, and tri-methylated lysines through alpha-ketoglutarate dependent reactions (Kooistra, S.M. and K. Helin, Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol, 2012. 13(5): p. 297-311; Mosammaparast, N. and Y. Shi, Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu Rev 20 Biochem, 2010. 79: p. 155-79).
  • Methylated histone marks on H3K4 and H3K9 are generally coupled with transcriptional activation and repression, respectively.
  • corepressor complexes e.g., CoREST
  • LSD1 has been reported to demethylate H3K4 and repress transcription
  • LSD1 in nuclear hormone receptor complex (e.g., androgen receptor), may demethylate H3K9 to activate gene 25 expression
  • LSD1 in nuclear hormone receptor complex
  • LSD1 androgen-receptor-dependent transcription. Nature, 2005. 437(7057): p. 436-9; Kahl, P., et al., Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res, 2006. 66(23): p. 11341- 7). This suggests the substrate specificity of LSD1 can be determined by associated factors, 30 thereby regulating alternative gene expressions in a context dependent manner. In addition to histone proteins, LSD1 may demethylate non-histone proteins. These include p53 (Huang, J., et
  • LSD1 also associates with other epigenetic regulators, such as DNA methyltransferase 1 (DNMT1) (Wang, J., et al., The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat Genet, 2009. 41(1): p. 125-9) and histone deacetylases (HDACs) complexes 15 (Hakimi, M.A., et al., A core-BRAF35 complex containing histone deacetylase mediates
  • DNMT1 DNA methyltransferase 1
  • HDACs histone deacetylases
  • LSD1 has been reported to contribute to a variety of biological processes, including cell proliferation, epithelial-mesenchymal transition (EMT), and stem cell biology (both embryonic stem cells and cancer stem cells) or self-renewal and cellular transformation of somatic cells (Chen, Y., et al., Lysine-specific histone demethylase 1 (LSD1): A potential molecular target for 30 tumor therapy. Crit Rev Eukaryot Gene Expr, 2012. 22(1): p. 53-9; Sun, G., et al., Histone
  • demethylase LSD1 regulates neural stem cell proliferation. Mol Cell Biol, 2010. 30(8): p. 1997-
  • AMLs Acute myeloid leukemias
  • LSC leukemia stem cell
  • LSD1 may regulate a subset of genes involved in multiple oncogenic programs to maintain LSC (Harris, W.J., et al., The histone demethylase KDM1A sustains the oncogenic potential of MLL- 20 AF9 leukemia stem cells. Cancer Cell, 2012. 21(4): p. 473-87; Schenk, T., et al., Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med, 2012. 18(4): p. 605-11). These findings suggest potential therapeutic benefit of LSD1 inhibitors targeting cancers having stem cell properties, such as AMLs.
  • LSD1 Overexpression of LSD1 is frequently observed in many types of cancers, including bladder cancer, NSCLC, breast carcinomas, ovary cancer, glioma, colorectal cancer, sarcoma including chondrosarcoma, Ewing’s sarcoma, osteosarcoma, and rhabdomyosarcoma, neuroblastoma, prostate cancer, esophageal squamous cell carcinoma, and papillary thyroid carcinoma.
  • studies found over-expression of LSD1 was significantly associated with 30 clinically aggressive cancers, for example, recurrent prostate cancer, NSCLC, glioma, breast, colon cancer, ovary cancer, esophageal squamous cell carcinoma, and neuroblastoma.
  • bladder cancer including bladder cancer, NSCLC, breast carcinomas, ovary cancer, glioma, colorectal cancer, sarcoma including chondrosarcoma, Ewing’s sarcoma, osteosarcoma
  • LSD1 Lysine-specific demethylase 1
  • CD86 expression is a marker of maturation of dendritic cells (DCs) which are
  • CD86 functions as a co-stimulatory factor to activate T cell proliferation (Greaves, P. and J.G. Gribben, The role of B7 family molecules in hematologic malignancy. Blood, 2013. 121(5): p. 734-44; Chen, L. and D.B. Flies, Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol, 2013. 13(4): p. 227- 5 42).
  • LSD1 activity has also been associated with viral pathogenesis.
  • LSD1 activity appears to be linked with viral replications and expressions of viral genes.
  • LSD1 functions as a co-activator to induce gene expression from the viral immediate early genes of various type of herpes virus including herpes 10 simplex virus (HSV), varicella zoster virus (VZV), and -herpesvirus human cytomegalovirus (Liang, Y., et al., Targeting the JMJD2 histone demethylases to epigenetically control herpesvirus infection and reactivation from latency. Sci Transl Med, 2013. 5(167): p.
  • HSV herpes 10 simplex virus
  • VZV varicella zoster virus
  • -herpesvirus human cytomegalovirus Liang, Y., et al., Targeting the JMJD2 histone demethylases to epigenetically control herpesvirus infection and reactivation from latency. Sci Trans
  • fetal globin gene would be potentially therapeutically beneficial for the disease of -globinopathies, including -thalassemia and sickle cell disease where the production of normal ⁇ -globin, a component of adult hemoglobin, is impaired (Sankaran, V.G. and S.H. Orkin, The switch from 25 fetal to adult hemoglobin. Cold Spring Harb Perspect Med, 2013. 3(1): p. a011643; Bauer, D.E., S.C. Kamran, and S.H. Orkin, Reawakening fetal hemoglobin: prospects for new therapies for the beta-globin disorders. Blood, 2012. 120(15): p. 2945-53).
  • LSD1 inhibition may potentiate other clinically used therapies, such as hydroxyurea or azacitidine. These agents may act, at least in part, by increasing -globin gene expression through different mechanisms.
  • LSD1 contributes to tumor development by altering epigenetic marks on histones and non-histone proteins. Accumulating data have validated that either genetic
  • LSD1 inhibitors alone or in combination with established therapeutic drugs would be effective to treat the diseases associated with LSD1 5 activity.
  • the present invention is directed to, inter alia, a compound of Formula I:
  • the present invention is further directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I and at least one pharmaceutically acceptable carrier.
  • the present invention is further directed to a method of inhibiting LSD1 comprising 15 contacting the LSD1 with a compound of Formula I.
  • the present invention is further directed to a method of treating an LSD1-mediated disease in a patient comprising administering to the patient a therapeutically effective amount of a compound of Formula I. 20 DETAILED DESCRIPTION
  • the present invention provides, inter alia, LSD1-inhibiting compounds such as a compound of Formula I:
  • X is N or CR X ;
  • Ring A is C6-10 aryl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C 6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from R A ;
  • Ring B is C6-10 aryl; 5-10 membered heteroaryl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; C3-10 cycloalkyl; or 4-10 membered heterocycloalkyl 10 comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S; wherein said C 6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from R B ;
  • R 1 is halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, Cy 1 , CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 ,
  • R 1 is not CN
  • R 2 is H, halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, Cy 2 , CN, OR a2 , SR a2 , 25 C(O)R b2 , C(O)NR c2 R d2 , C(O)OR a2 , OC(O)R b2 , OC(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 ,
  • each R B is independently selected from Cy 3 , halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 1-6 haloalkyl, CN, NO 2 , OR a5 , SR a5 , C(O)R b5 , C(O)NR c5 R d5 , C(O)OR a5 , OC(O)R b5 ,
  • NR c7 C( NR e7 )NR c7 R d7 , NR c7 S(O)R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , S(O)R b7 ,
  • each Cy 1 , Cy 2 , Cy 3 , and Cy 4 is independently selected from C 6-10 aryl, C 3-10 cycloalkyl, 5- 10 membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally 5 substituted with 1, 2, 3, or 4 substituents independently selected from R Cy ;
  • NR c6 C( NR e6 )NR c6 R d6 , NR c6 R d6 , NR c6 C(O)R b6 , NR c6 C(O)OR a6 , NR c6 C(O)NR c6 R d6 ,
  • each R a1 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and Cy 4 ; wherein said C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy 4 , halo, CN, OR a3 , SR a3 , C(O)R b3 , C(O)NR c3 R d3 , C(O)OR a3 , OC(O)R b3 , OC(O)NR c3 R d3 , NR c3 R d3 , NR c3 C(O)R b3 , NR c3 C(O)OR a3 ,
  • each R b1 , R c1 , and R d1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)- 30 C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6
  • heterocycloalkyl C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 cyanoalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , 5 NR c7 C(O)R b7 , NR c7 C(O)NR c7 R d7 , NR c7 C
  • R c1 and R d1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)NR c7 R d7 , NR c7 C(O)NR c7 R d7
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , and S(O) 2 NR c7 R d7 ;
  • each R a2 , R b2 , R c2 , and R d2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25 heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10
  • R c2 and R d2 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 ,
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O) 2 R b7 , NR c7 S(O) 2 R b7 , NR c7 S(O) 2 NR c7 R d7 , and S(O) 2 NR c7 R d7 ;
  • each R a3 , R b3 , R c3 , and R d3 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, 15 C 2-6 alkenyl, C 2-6 alkynyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10
  • R c3 and R d3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 ,
  • each R a4 , R b4 , R c4 , and R d4 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C 1-4 alkyl, C 1- 10 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 ,
  • R c4 and R d4 together with the N atom to which they are attached form a 4-, 5-, 6-, 15 or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
  • each R a5 , R b5 , R c5 , and R d5 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25 heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10
  • R c5 and R d5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , 5 C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)NR c7 R d7 ,
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O) 2 R b7 , NR c7 S(O) 2 R b7 , NR c7 S(O) 2 NR c7 R d7 , and S(O) 2 NR c7 R d7 ;
  • each R a6 , R b6 , R c6 , and R d6 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, 15 C 2-6 alkenyl, and C 2-6 alkynyl, wherein said C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C 1-4 alkyl, C 1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 ,
  • R c6 and R d6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 ,
  • each R a7 , R b7 , R c7 , and R d7 is independently selected from H, C1-4 alkyl, C1-4 haloalkyl, C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, 30 halo, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, C1-4 alkylamino, di(C1-4 alkyl)amino, C1-4 haloalkyl, and C1-4 haloalkoxy; and
  • each R e1 , R e2 , R e3 , R e4 , R e5 , R e6 , and R e7 is independently selected from H, C1-4 alkyl, and CN. In some embodiments:
  • 5 X is N or CR X ;
  • Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from R A ;
  • Ring B is phenyl or 5-6 membered heteroaryl comprising carbon and 1, 2, 3 or 4
  • R 1 is halo, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, Cy 1 , CN, OR a1 , SR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , OC(O)R b1 , OC(O)NR c1 R d1 , NR c1 R d1 , NR c1 C(O)R b1 ,
  • R 1 is not CN
  • R 2 is H, halo, C1-6 alkyl, CN, OR a2 , C(O)R b2 , C(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 , S(O)2R b2 , or S(O)2NR c2 R d2 ; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 25 substituents independently selected from halo, CN, OR a2 , C(O)R b2 , C(O)NR c2 R d2 , NR c2 R d2 , NR c2 C(O)R b2 , S(O) 2 R b2 , and S(O) 2 NR c2 R d2 ;
  • each R A is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, OR a4 , C(O)R b4 , C(O)NR c4 R d4 , C(O)OR a4 , NR c4 R d4 , NR c4 C(O)R b4 , S(O)2R b4 , and S(O)2NR c4 R d4 , wherein said C 1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected 30 from halo, C d4
  • each R B is independently selected from Cy 3 , halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 5
  • R X is independently selected from H, halo, C1-6 alkyl, C1-6 haloalkyl, CN, OR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , NR c7 R d7 , NR c7 C(O)R b7 , S(O)2R b7 , and S(O)2NR c7 R d7 ;
  • each Cy 1 , Cy 3 , and Cy 4 is independently selected from C 6-10 aryl, C 3-10 cycloalkyl, 5-10 10 membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally
  • each R Cy is independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 cyanoalkyl, C 2-6 alkenyl, C 2-6 alkynyl, phenyl, C 3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 15 alkyl-, and (4-7 membered heterocycloalkyl)-C a6
  • NR c6 C( NR e6 )NR c6 R d6 , NR c6 R d6 , NR c6 C(O)R b6 , NR c6 C(O)OR a6 , NR c6 C(O)NR c6 R d6 ,
  • each R a1 is independently selected from H, C1-6 alkyl, and Cy 4 ; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy 4 , halo, CN, OR a3 , C(O)R b3 , C(O)NR c3 R d3 , C(O)OR a3 , NR c3 R d3 , NR c3 C(O)R b3 , S(O) 2 R b3 , and S(O) 2 NR c3 R d3 ; 30 each R b1 , R c1 , and R d1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered
  • heterocycloalkyl C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 5 C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 cyan
  • R c1 and R d1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C 6-10 aryl, 5-6 membered heteroaryl, C 1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)NR c7 R d7 , NR c7 C(O)NR c7 R d7
  • C 1-6 alkyl, C 3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C 1-4 cyanoalkyl, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , 20 NR c7 R d7 , NR c7 C(O)R b7 ,
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , and S(O) 2 NR c7 R d7 ;
  • each R a2 , R b2 , R c2 , and R d2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25 heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)- C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , 15 and S(O) 7
  • each R a3 , R b3 , R c3 , and R d3 is independently selected from H, C 1-6 alkyl, C 1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 2-6 20 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C
  • R c3 and R d3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C 1-6 alkyl, C 3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 30 5-6 membered heteroaryl, C 7
  • 1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c R d7 ,
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , and S(O) 7
  • each R a4 , R b4 , R c4 , and R d4 is independently selected from H, C 1-6 alkyl, C 1-4 haloalkyl, 10 C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C 1-4 cyanoalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 ,
  • R c4 and R d4 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 ,
  • each R a5 , R b5 , R c5 , and R d5 is independently selected from H, C 1-6 alkyl, C 1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 25 C 1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 2-6
  • R c5 and R d5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
  • NR c7 C( NR e7 )NR c7 R d7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 , NR c7 S(O)2NR c7 R d7 , 15 and S(O) 2 NR c7 R d7 ;
  • each R a6 , R b6 , R c6 , and R d6 is independently selected from H, C 1-6 alkyl, C 1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C 1-4 cyanoalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 ,
  • R c6 and R d6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
  • each R a7 , R b7 , R c7 , and R d7 is independently selected from H, C 1-4 alkyl, C 1-4 haloalkyl, 30 C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino,
  • each R e1 , R e6 , and R e7 is independently selected from H, C 1-4 alkyl, and CN. 5 In some embodiments:
  • X is N or CR X ;
  • Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from R A ;
  • Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected 10 from R B ;
  • R 1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy 1 , OR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , S(O) 2 R b1 , or S(O) 2 NR c1 R d1 ; wherein said C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy 1 , halo, CN, OR a1 , C(O)R b1 , C(O)NR c1 R d1 , 15 C(O)OR a1 , NR c1 R
  • R 2 is H
  • each R A is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, and OR a4 , wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from CN and OR a4 ;
  • each R B is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, and OR a5 ;
  • R X is H
  • each Cy 1 and Cy 4 is independently selected from phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 25 3, or 4 substituents independently selected from R Cy ;
  • each R Cy is independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, CN, NO2, OR a6 , SR a6 , C(O)R b6 , C(O)NR c6 R d6 , C(O)OR a6 , OC(O)R b6 , OC(O)NR c6 R d6 , NR c6 R d6 , NR c6 C(O)R b6 , NR c6 C(O)OR a6 , NR c6 C(O)NR c6 R d6 , NR c6 S(O)R b6 , NR c6 S(O) 2 R b6 , NR c6 S(O) 2 NR c6 R d6 , S(O)R b6
  • each R a1 is independently selected from H, C1-6 alkyl, and 4-7 membered
  • each R b1 , R c1 , and R d1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C 2-6 alkynyl, phenyl, C 3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C 1-4 alkyl-, C 3-7 cycloalkyl-C 1-4 alkyl-, (5-6 membered heteroaryl)-C 1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2- 15 6 alkynyl, phenyl, C 3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C 1-4 alkyl-, C 3-7 cycloalkyl-C 1-4 alkyl
  • R c1 and R d1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 25 aryl, 5-6 membered heteroaryl, C 1-6 haloalkyl, halo, CN, OR a7 , SR a7 , C(O)R b7 , C(O)NR c7 R d7 , C(O)OR a7 , OC(O)R b7 , OC(O)NR c7 R d7 , NR c7 R d7 , NR c7 C(O)R b7 , NR c7 C(O)NR c7 R d7 ,
  • NR c7 C(O)NR c7 R d7 NR c7 C(O)OR a7 , S(O)R b7 , S(O)NR c7 R d7 , S(O)2R b7 , NR c7 S(O)2R b7 ,
  • each R a5 is independently selected from H and C 1-6 alkyl
  • each R a6 , R b6 , R c6 , and R d6 is independently selected from H and C 1-6 alkyl; and each R a7 , R b7 , R c7 , and R d7 is independently selected from H and C1-4 alkyl.
  • X is N.
  • X is CR X .
  • Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C 6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from R A .
  • Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from R A .
  • Ring A is phenyl substituted by one R A .
  • Ring A is phenyl substituted by CN.
  • Ring B is phenyl or 5-6 membered heteroaryl comprising carbon 20 and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; wherein said phenyl and 5-6 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from R B .
  • Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected from R B .
  • Ring B is phenyl substituted by one R B .
  • Ring B is phenyl substituted by methyl.
  • R 1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy 1 , OR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , NR c1 R d1 , NR c1 C(O)R b1 , NR c1 C(O)OR a1 , S(O) 2 R b1 , or S(O) 2 NR c1 R d1 ; wherein said C 1-6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each 30 optionally substituted with 1, 2, or 3 substituents independently selected from Cy 1 , halo, CN, OR a1 , C(O)R b1 , C(O)NR c1 R d1 , C(O)OR a1 , NR
  • R 1 is C1-6 alkyl, Cy 1 , or OR a1 , wherein said C1-6 alkyl is substituted with one Cy 1 .
  • R 1 is pyrrolidin-3-ylmethoxy, 2-pyrrolidin-3-ylethyl, (1- methylpyrrolidin-3-yl)ethyl, 3-[(methylamino)methyl]phenyl, 3-aminopyrrolidin-1- 5 yl)methyl]phenyl, piperazin-1-ylmethyl, 4-methylpiperazin-1-yl)methyl, 3- (dimethylamino)pyrrolidin-1-yl, 3-(methylamino)pyrrolidin-1-yl, or (1-methylpyrrolidin-3- yl)methoxy.
  • R 2 is H.
  • each R A is independently selected from halo, C 1-6 alkyl, C 1-6 10 haloalkyl, CN, OR a4 , C(O)R b4 , C(O)NR c4 R d4 , C(O)OR a4 , NR c4 R d4 , NR c4 C(O)R b4 , S(O) b4
  • C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from halo, C 1-6 alkyl, C 1-6 haloalkyl, CN, OR a4 , C(O)R b4 , C(O)NR c4 R d4 , C(O)OR a4 , NR c4 R d4 , NR c4 C(O)R b4 , S(O) 2 R b4 , and S(O) 2 NR c4 R d4 .
  • each R A is independently selected from halo, C1-6 alkyl, C1-6 15 haloalkyl, CN, and OR a4 , wherein said C 1-6 alkyl is optionally substituted by 1, 2, or 3,
  • R A is CN
  • each R B is independently selected from Cy 3 , halo, C1-6 alkyl, C2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, CN, NO 2 , OR a5 , C(O)R b5 , C(O)NR c5 R d5 , C(O)OR a5 , 20 NR c5 R d5 , NR c5 C(O)R b5 , S(O)2R b5 , and S(O)2NR c5 R d5 , wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3 substituents independently selected from Cy 3 , halo, C 1-6 haloalkyl, CN, NO 2 , OR a5 , C(O)R b5 , C(O)NR c5 R d5 , C(O)
  • each R B is independently selected from halo, C1-6 alkyl, C2-6 25 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, CN, and OR a5 .
  • R B is C 1-6 alkyl.
  • R B is methyl
  • R X is H.
  • each Cy 1 is independently selected from phenyl, C 3-7 cycloalkyl, 30 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R Cy .
  • each Cy 1 is phenyl or 4-7 membered heterocycloalkyl, each optionally substituted with 1 or 2 substituents independently selected from R Cy .
  • each Cy 1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from R Cy .
  • each Cy 1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from C 1-4 alkyl and NR c6 R d6 , wherein said C1-4 alkyl is optionally substituted with NR c6 R d6 .
  • each R Cy is C1-4 alkyl and NR c6 R d6 , wherein said C1-4 alkyl is optionally substituted with NR c6 R d6 .
  • each R a1 is independently selected from H, C1-6 alkyl, and Cy 4 ; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy 4 , halo, CN, OR a3 , C(O)R b3 , C(O)NR c3 R d3 , C(O)OR a3 , NR c3 R d3 , NR c3 C(O)R b3 , S(O) 2 R b3 , and S(O) 2 NR c3 R d3 .
  • each R a1 is independently selected from H, C1-6 alkyl, and 4-7 15 membered heterocycloalkyl; wherein said C 1-6 alkyl is optionally substituted with 1, 2, or 3
  • each R a1 is C1-4 alkyl substituted by 4-7 membered
  • heterocycloalkyl wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, OR a6 , C(O)R b6 , C(O)NR c6 R d6 , C(O)OR a6 , NR c6 R d6 , NR c6 C(O)R b6 , S(O)2R b6 , and
  • each R a1 is pyrrolidinylmethyl optionally substituted with one C 1-4 alkyl.
  • the compounds of the invention have Formula IIa:
  • the compounds of the invention have Formula IIb: 5
  • the compounds of the invention have Formula IIIa:
  • the compounds of the invention have Formula IIIb:
  • the phrase "optionally substituted” means unsubstituted or substituted.
  • substituted means that a hydrogen atom is removed and replaced by a monovalent substituent, or two hydrogen atoms are replaced with a divalent substituent like a terminal oxo group. It is to be understood that substitution at a given atom is limited by valency.
  • C i-j indicates a range which includes the endpoints, wherein i and j are integers and indicate the number of carbons. Examples include C1-4, C1-6, and 15 the like.
  • z-membered typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is z.
  • piperidinyl is an example of a 6-membered heterocycloalkyl ring
  • pyrazolyl is an example of a 5- membered heteroaryl ring
  • pyridyl is an example of a 6-membered heteroaryl ring
  • 1, 2, 3, 4- 20 tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
  • Ci-j alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched, having i to j carbons.
  • the alkyl group contains from 1 to 6 carbon atoms or from 1 to 4 carbon atoms, or from 1 to 3 carbon atoms.
  • alkyl moieties include, but are not25 limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, and t- butyl.
  • Ci-j alkoxy employed alone or in combination with other terms, refers to a group of formula -O-alkyl, wherein the alkyl group has i to j carbons.
  • Example alkoxy groups include methoxy, ethoxy, and propoxy (e.g., n-propoxy and isopropoxy).
  • the alkyl group has 1 to 3 carbon atoms.
  • C i-j alkenyl employed alone or in combination with other terms, refers to an unsaturated hydrocarbon group having one or more double carbon-carbon bonds and having i to j carbons.
  • the alkenyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
  • Example alkenyl groups include, but are not limited to, ethenyl, n-propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.
  • Ci-j alkynyl employed alone or in combination with other terms, refers to an unsaturated hydrocarbon group having one or more triple carbon-carbon bonds and having i to j carbons.
  • Example alkynyl groups include, but are not limited to, ethynyl, propyn-1-yl, propyn-2-yl, and the like.
  • the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
  • C i-j alkylamino refers to a group of formula -NH(alkyl), wherein the alkyl group has i to j carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. In some embodiments, the alkylamino group is–NH(C1-4 alkyl) such as, for example, methylamino, ethylamino or propylamino.
  • C i-j alkylthio employed alone or in combination with other terms, refers to a group of formula -S-alkyl, wherein the alkyl group has i to j carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. In some embodiments, the alkylthio group is C1-4 alkylthio such as, for example, methylthio or ethylthio.
  • amino employed alone or in combination with other terms, 30 refers to a group of formula–NH2.
  • aryl employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon, such as, but not limited to, phenyl, 1-naphthyl, 2-naphthyl, anthracenyl, phenanthrenyl, and the like.
  • aryl is C6-10 aryl.
  • the aryl group is a
  • the aryl group is phenyl
  • aryl-C i-j alkyl employed alone or in combination with other terms, refers to an alkyl group substituted by an aryl group.
  • An example of a aryl-Ci-j alkyl group is benzyl.
  • carbonyl employed alone or in combination with other terms, 10 refers to a -C(O)- group.
  • Cycloalkyl refers to a non-aromatic cyclic hydrocarbon moiety having i to j ring-forming carbon atoms, which may optionally contain one or more alkenylene groups as part of the ring structure.
  • Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) ring 15 systems. Also included in the definition of cycloalkyl are moieties that have one or more
  • cycloalkyl is C 3-10 cycloalkyl, C 3-7 cycloalkyl, or C 5-6 cycloalkyl.
  • Exemplary 20 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl,
  • cyclopentenyl cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, and the like.
  • Further exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • Ci-j cycloalkyl-Ci-j alkyl employed alone or in combination 25 with other terms, refers to an alkyl group substituted by a cycloalkyl group.
  • An example of a C i-j cycloalkyl-C i-j alkyl group is cyclopropylmethyl.
  • Ci-j haloalkoxy refers to a group of formula–O-haloalkyl having i to j carbon atoms.
  • An example haloalkoxy group is OCF 3 .
  • An additional example haloalkoxy group is OCHF 2 .
  • the 30 haloalkoxy group is fluorinated only.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • the haloalkoxy group is C1-4 haloalkoxy.
  • halo refers to a halogen atom selected from F, Cl, I or Br. In some embodiments, "halo" refers to a halogen atom selected from F, Cl, or Br. In some embodiments, the halo substituent is F.
  • Ci-j haloalkyl refers to an alkyl group having from one halogen atom to 2s+1 halogen atoms which may be the same or different, where "s" is the number of carbon atoms in the alkyl group, wherein the alkyl group has i to j carbon atoms.
  • the haloalkyl group is fluorinated only.
  • the haloalkyl group is fluoromethyl, difluoromethyl, or
  • the haloalkyl group is trifluoromethyl. In some embodiments, the haloalkyl group is trifluoromethyl. In some
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • heteroaryl employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic heterocylic moiety, having one or more heteroatom ring members selected from nitrogen, sulfur and oxygen.
  • the heteroaryl group has 1, 2, 3, or 4 heteroatom ring 15 members.
  • the heteroaryl group has 1, 2, or 3 heteroatom ring members.
  • the heteroaryl group has 1 or 2 heteroatom ring members. In some embodiments, the heteroaryl group has 1 heteroatom ring member. In some embodiments, the heteroaryl group is 5- to 10-membered or 5- to 6-membered. In some embodiments, the heteroaryl group is 5-membered. In some embodiments, the heteroaryl group is 6-membered. 20 When the heteroaryl group contains more than one heteroatom ring member, the heteroatoms may be the same or different. The nitrogen atoms in the ring(s) of the heteroaryl group can be oxidized to form N-oxides.
  • Example heteroaryl groups include, but are not limited to, pyridine, pyrimidine, pyrazine, pyridazine, pyrrole, pyrazole, azolyl, oxazole, isoxazole, thiazole, isothiazole, imidazole, furan, thiophene, triazole, tetrazole, thiadiazole, quinoline, isoquinoline, 25 indole, benzothiophene, benzofuran, benzisoxazole, imidazo[1, 2-b]thiazole, purine, triazine, and the like.
  • a 5-membered heteroaryl is a heteroaryl group having five ring-forming atoms comprising wherein one or more of the ring-forming atoms are independently selected from N, O, and S.
  • the 5-membered heteroaryl group has 1, 2, or 3 heteroatom ring 30 members.
  • the 5-membered heteroaryl group has 1 or 2 heteroatom ring members.
  • the 5-membered heteroaryl group has 1 heteroatom ring
  • Example ring-forming members include CH, N, NH, O, and S.
  • Example five- membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1, 2, 3-triazolyl, tetrazolyl, 1, 2, 3-thiadiazolyl, 1, 2, 3-oxadiazolyl, 1, 2, 4-triazolyl, 1, 2, 4-thiadiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-triazolyl, 1, 3, 4-thiadiazolyl, and 1, 3, 5 4-oxadiazolyl.
  • a 6-membered heteroaryl is a heteroaryl group having six ring-forming atoms wherein one or more of the ring-forming atoms is N.
  • the 6-membered heteroaryl group has 1, 2, or 3 heteroatom ring members.
  • the 6-membered heteroaryl group has 1 or 2 heteroatom ring members.
  • the 6-membered 10 heteroaryl group has 1 heteroatom ring member.
  • Example ring-forming members include CH and N.
  • Example six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl, and pyridazinyl.
  • heteroaryl-C i-j alkyl employed alone or in combination with other terms, refers to an alkyl group substituted by a heteroaryl group.
  • heteroaryl-C i-j alkyl employed alone or in combination with other terms, refers to an alkyl group substituted by a heteroaryl group.
  • heteroaryl-C i-j alkyl group is pyridylmethyl.
  • heterocycloalkyl refers to non-aromatic heterocyclic ring system, which may optionally contain one or more unsaturations as part of the ring structure, and which has at least one heteroatom ring member independently selected from nitrogen, sulfur and oxygen.
  • the 20 heterocycloalkyl group has 1, 2, 3, or 4 heteroatom ring members.
  • the heterocycloalkyl group has 1, 2, or 3 heteroatom ring members.
  • the heterocycloalkyl group has 1 or 2 heteroatom ring members.
  • the heterocycloalkyl group has 1 heteroatom ring member.
  • Example ring-forming members include CH, CH 2 , C(O), N, NH, O, S, S(O), and S(O) 2 .
  • Heterocycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) ring systems, including spiro systems. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the non- aromatic ring, for example, 1, 2, 3, 4-tetrahydro-quinoline, dihydrobenzofuran and the like.
  • the 30 carbon atoms or heteroatoms in the ring(s) of the heterocycloalkyl group can be oxidized to form a carbonyl, sulfinyl, or sulfonyl group (or other oxidized linkage) or a nitrogen atom can be
  • the heterocycloalkyl is 5- to 10-membered, 4- to 10- membered, 4- to 7-membered, 5-membered, or 6-membered.
  • heterocycloalkyl groups include 1, 2, 3, 4-tetrahydro-quinoline, dihydrobenzofuran, azetidine, azepane, pyrrolidine, piperidine, piperazine, morpholine, thiomorpholine, and pyran.
  • heterocycloalkyl-C i-j alkyl refers to an alkyl group substituted by a heterocycloalkyl group.
  • An example of a heterocycloalkyl-Ci-j alkyl group is pyrrolidinylmethyl.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereoisomers, are intended unless 10 otherwise indicated.
  • the compounds of the invention can be any of the possible stereoisomers.
  • the stereochemistry of 20 the chiral center can be (R) or (S).
  • the stereochemistry of the chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R) and (R), (R) and (S); (S) and (R), or (S) and (S).
  • each of the three chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R), (R) and (R); (R), (R) and (S); (R), (S) and 25 (R); (R), (S) and (S); (S), (R) and (R); (S), (R) and (S); (S), (S) and (R); or (S), (S) and (S).
  • An example method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and 30 L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as -camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of -methylbenzylamine (e.g., S and R forms, or diastereoisomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1, 2- diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant 10 migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric
  • Example prototropic tautomers include ketone– enol pairs, amide - imidic acid pairs, lactam– lactim pairs, amide - imidic acid pairs, enamine– imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 15 1, 2, 4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • All compounds, and pharmaceutically acceptable salts thereof, can be found together 25 with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated.
  • the compounds of the invention, or salts thereof are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in a compound of the invention. 30
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound 5 medical judgment, suitable for use in contact with the tissues of human beings and animals
  • ambient temperature and “room temperature,” as used herein, are understood in the art, and refer generally to a temperature, e.g., a reaction temperature, that is 10 about the temperature of the room in which the reaction is carried out, for example, a
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or 15 base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of 20 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, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or 25 butanol) or acetonitrile (MeCN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or 25 butanol) or acetonitrile (MeCN) are preferred.
  • suitable salts are found in Remington's Pharmaceutical Sciences, 17 th Ed., (Mack Publishing Company, Easton, 1985), p. 1418, Berge et al., J. Pharm. Sci., 1977, 66(1), 1-19, and in Stahl et al
  • Suitable solvents can be substantially non-reactive with the starting materials (reactants), the
  • reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • Preparation of compounds of the invention can involve the protection and deprotection of 30 various chemical groups. The need for protection and deprotection, and the selection of
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV- visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectroscopy (LCMS), or thin layer chromatography (TLC).
  • HPLC high performance liquid chromatography
  • LCMS liquid chromatography-mass spectroscopy
  • TLC thin layer chromatography
  • Compounds can be purified by those skilled in the art by a variety of 10 methods, including high performance liquid chromatography (HPLC) ("Preparative LC-MS Purification: Improved Compound Specific Method Optimization" Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs J. Combi. Chem. 2004, 6(6), 874-883, which is incorporated here
  • Compounds of formula 9 can be synthesized as shown in Scheme 1.
  • Compound 1 can 15 undergo Suzuki reaction with an appropriate boronic acid or ester of formula 2 in the presence of a palladium catalyst and a suitable base such as K 2 CO 3 to provide compound of formula 3.
  • ring B to give compound of formula 5 can be achieved by coupling of compound 3 with compound of formula 4 under standard Suzuki coupling conditions (M is a boronic acid or ester, with palladium catalysis), or standard Negishi coupling conditions (M is Zn-halo, in the 20 presence of a palladium catalyst), or standard Buchwald amination conditions (M is H attached to a ring-forming N atom in ring B, in the presence of a palladium catalyst and a suitable base).
  • Halogenation of compound 5 using N-chlorosuccinimide, N-bromosuccinimide or N- iodosuccinimide can provide a compound of formula 6 (Hal is Cl, Br or I).
  • Compound 6 can be converted to a formamidoxime derivative of formula 7 by reacting with N,N-dimethylformamide 25 dimethyl acetal, followed by treatment with hydroxylamine.
  • the formamidoxime derivative 7 can undergo cyclization upon treating with trifluoroacetic anhydride (TFAA) to afford a triazole compound of formula 8.
  • TFAA trifluoroacetic anhydride
  • the aryl halide 8 can react with R 1 -M to give a compound of formula 9 under standard cross coupling conditions, such as Suzuki coupling conditions (M is a boronic acid or ester, with palladium catalysis), Sonogashira coupling conditions (M is a
  • R 1 -M is an alcohol (M is H), with palladium or copper catalysis).
  • compounds of formula 11 can be prepared as shown in Scheme 3.
  • Aryl halide 6 can react with R 1 -M under standard cross coupling conditions as described in Scheme 1 (e.g., Suzuki coupling, Negishi coupling, Sonogashira coupling, Buchwald amination or Ullmann coupling) to give compounds of formula 12.
  • Condensation of amino-pyridine derivative 12 with ethoxycarbonyl isothiocyanate, followed by treatment with hydroxylamine can give the
  • Transformation of the amino group in compound 13 to bromide can be achieved under standard Sandmeyer reaction conditions (e.g., NaNO 2 , HBr then CuBr) to give compounds of formula 14.
  • Functionalization of aryl bromide 14 with an R 2 substituent to give compound 11 can be performed under standard cross coupling reaction conditions (e.g., Suzuki 5 coupling, Negishi coupling, Sonogashira coupling, Buchwald amination or Ullmann coupling) as described in the previous Schemes.
  • Compounds of the invention are LSD1 inhibitors and, thus, are useful in treating diseases and disorders associated with activity of LSD1.
  • any of the compounds of the invention including any of the embodiments thereof, may be used.
  • the compounds of the invention are selective for LSD1 over 15 LSD2, meaning that the compounds bind to or inhibit LSD1 with greater affinity or potency, compared to LSD2.
  • selectivity can be at least about 5-fold, at least about 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold.
  • LSD1-mediated disease As inhibitors of LSD1, the compounds of the invention are useful in treating LSD1- 20 mediated diseases and disorders.
  • LSD1-mediated disease or“LSD1-mediated
  • LSD1 refers to any disease or condition in which LSD1 plays a role, or where the disease or condition is associated with expression or activity of LSD1.
  • the compounds of the invention can be any disease or condition in which LSD1 plays a role, or where the disease or condition is associated with expression or activity of LSD1.
  • Diseases and conditions treatable using the compounds of the invention include generally cancers, inflammation, autoimmune diseases, viral induced pathogenesis, beta-globinopathies, 5 and other diseases linked to LSD1 activity.
  • Cancers treatable using compounds according to the present invention include, for example, hematological cancers, sarcomas, lung cancers, gastrointestinal cancers, genitourinary tract cancers, liver cancers, bone cancers, nervous system cancers, gynecological cancers, and skin cancers.
  • Example hematological cancers include, for example, lymphomas and leukemias such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma,
  • ALL acute lymphoblastic leukemia
  • AML acute myelogenous leukemia
  • APL acute promyelocytic leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • DLBCL diffuse large B-cell lymphoma
  • Non-Hodgkin lymphoma including relapsed or refractory NHL and recurrent
  • myeloproliferative diseases e.g., primary myelofibrosis (PMF), polycythemia vera (PV),
  • PMF primary myelofibrosis
  • PV polycythemia vera
  • E essential thrombocytosis
  • MDS myelodysplasia syndrome
  • multiple myeloma multiple myeloma
  • Example sarcomas include, for example, chondrosarcoma, Ewing’s sarcoma, osteosarcoma, rhabdomyosarcoma, angiosarcoma, fibrosarcoma, liposarcoma, myxoma, rhabdomyoma, fibroma, lipoma, harmatoma, and teratoma.
  • Example lung cancers include, for example, non-small cell lung cancer (NSCLC),
  • bronchogenic carcinoma squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma
  • alveolar carcinoma bronchial adenoma
  • chondromatous hamartoma chondromatous hamartoma
  • mesothelioma squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma
  • alveolar carcinoma bronchiolar carcinoma
  • bronchial adenoma chondromatous hamartoma
  • mesothelioma mesothelioma
  • Example gastrointestinal cancers include, for example, cancers of the esophagus 25 (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), and 30 colorectal cancer.
  • Example genitourinary tract cancers include, for example, cancers of the kidney
  • bladder and urethra squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), and testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, 5 sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma).
  • Example liver cancers include, for example, hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.
  • hepatoma hepatocellular carcinoma
  • cholangiocarcinoma hepatoblastoma
  • angiosarcoma hepatocellular adenoma
  • hemangioma hemangioma
  • Example bone cancers include, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant 10 lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma, and giant cell tumors
  • osteogenic sarcoma osteosarcoma
  • fibrosarcoma malignant fibrous histiocytoma
  • chondrosarcoma chondrosarcoma
  • Ewing's sarcoma malignant 10 lymphoma
  • multiple myeloma malignant giant cell tumor chordoma
  • osteochronfroma osteocar
  • Example nervous system cancers include, for example, cancers of the skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma,
  • meningiosarcoma 15 meningiosarcoma, gliomatosis), brain (astrocytoma, meduoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma,
  • retinoblastoma retinoblastoma, congenital tumors), and spinal cord (neurofibroma, meningioma, glioma, sarcoma), as well as neuroblastoma and Lhermitte-Duclos disease.
  • Example gynecological cancers include, for example, cancers of the uterus (endometrial 20 carcinoma), cervix (cervical carcinoma, pre -tumor cervical dysplasia), ovaries (ovarian
  • carcinoma serotonin-associated cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma
  • granulosa-thecal cell tumors Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma
  • vulva squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma
  • vagina vagina
  • Example skin cancers include, for example, melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, and keloids.
  • the compounds of the invention can further be used to treat cancer types where LSD1 30 may be overexpressed including, for example, breast, prostate, head and neck, laryngeal, oral, and thyroid cancers (e.g., papillary thyroid carcinoma).
  • cancer types where LSD1 30 may be overexpressed including, for example, breast, prostate, head and neck, laryngeal, oral, and thyroid cancers (e.g., papillary thyroid carcinoma).
  • the compounds of the invention can further be used to treat genetic disorders such as Cowden syndrome and Bannayan-Zonana syndrome.
  • the compounds of the invention can further be used to treat viral diseases such as herpes simplex virus (HSV), varicella zoster virus (VZV), human cytomegalovirus, hepatitis B virus 5 (HBV), and adenovirus.
  • viral diseases such as herpes simplex virus (HSV), varicella zoster virus (VZV), human cytomegalovirus, hepatitis B virus 5 (HBV), and adenovirus.
  • the compounds of the invention can further be used to treat beta-globinopathies including, for example, beta-thalassemia and sickle cell anemia.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • "contacting" a LSD1 protein with a 10 compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a LSD1 protein, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the LSD1 protein.
  • cattle, sheep, horses, or primates and most preferably humans.
  • the phrase "therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, 20 medical doctor or other clinician.
  • treating refers to inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e.,, arresting further development of the pathology and/or symptomatology) or ameliorating the disease; for 25 example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e.,, reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • preventing refers to preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to 30 the disease, condition or disorder but does not yet experience or display the pathology or
  • the compounds of the invention can be used in combination treatments where the compound of the invention is administered in conjunction with other treatments such as the 5 administration of one or more additional therapeutic agents.
  • the additional therapeutic agents are typically those which are normally used to treat the particular condition to be treated.
  • the additional therapeutic agents can include, e.g., chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF, FAK, JAK, PIM, PI3K inhibitors for treatment of LSD1-mediated diseases, disorders or conditions.
  • the one or more additional 10 pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • the compounds of the invention can be used in combination with a therapeutic agent that targets an epigenetic regulator.
  • epigenetic regulators include the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases.
  • Histone deacetylase 15 inhibitors include, e.g., vorinostat.
  • the compounds of the invention can be used in combination with chemotherapeutic agents, agonists or antagonists of nuclear receptors, or other anti-proliferative agents.
  • the compounds of the invention can also be used in combination with medical therapy such as surgery or radiotherapy, e.g., gamma-radiation, 20 neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and
  • chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin,
  • cytarabine dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone
  • propionate eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, 30 gefitinib, gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate,
  • ibritumomab tiuxetan idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan,
  • lapatinib ditosylate lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone phenpropionate, nelarabine, nofetumomab, oxaliplatin, paclitaxel, pamidronate, panitumumab, pegaspargase, pegfilgrastim, 5 pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine,
  • rasburicase rituximab, ruxolitinib, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, teniposide, testolactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, vorinostat, and zoledronate.
  • the compounds of the invention can be used in combination with ruxolitinib.
  • the compound of the invention can be administered in combination with a corticosteroid such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • a corticosteroid such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
  • the compound of the invention can be administered in combination with an immune suppressant such as fluocinolone acetonide (Retisert®), rimexolone (AL-2178, Vexol, Alcon), or cyclosporine (Restasis®).
  • an immune suppressant such as fluocinolone acetonide (Retisert®), rimexolone (AL-2178, Vexol, Alcon), or cyclosporine (Restasis®).
  • the compound of the invention can be administered in combination with one or more additional agents selected from DehydrexTM 20 (Holles Labs), Civamide (Opko), sodium hyaluronate (Vismed, Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrinTM (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline25 (Duramycin, MOLI1901, Lantibio), CF101 (2S, 3S, 4R, 5R)
  • the compound of the invention can be administered in combination with one or more additional agents such as Hydrea® (hydroxyurea).
  • the compound of the invention can be administered in combination with one or more agents selected from an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and non-steroidal anti-inflammatories, and anti- 10 allergic agents.
  • agents selected from an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and non-steroidal anti-inflammatories, and anti- 10 allergic agents selected from an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and non-steroidal anti-inflammatories, and anti- 10 allergic agents.
  • suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin;
  • rifampins colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins"); 15 cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac; flurbiprofen; ketorolac; suprofen;
  • agents one or more of which a provided compound may also be combined with include: a treatment for Alzheimer's Disease such as donepezil and rivastigmine; 20 a treatment for Parkinson's Disease such as L-DOPA/carbidopa, entacapone, ropinirole,
  • pramipexole bromocriptine, pergolide, trihexyphenidyl, and amantadine
  • an agent for treating multiple sclerosis such as beta interferon (e.g., Avonex® and Rebif®), glatiramer acetate, and mitoxantrone
  • a treatment for asthma such as albuterol and montelukast
  • an agent for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol
  • an anti-inflammatory agent 25 such as a corticosteroid, such as dexamethasone or prednisone, a TNF blocker, IL-1 RA,
  • an immunomodulatory agent including immunosuppressive agents, such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, an interferon, a corticosteroid, cyclophosphamide, azathioprine, and sulfasalazine; a neurotrophic factor such as an acetylcholinesterase inhibitor, an MAO inhibitor, an interferon, an anti- 30 convulsant, an ion channel blocker, riluzole, or an anti-Parkinson's agent; an agent for treating cardiovascular disease such as a beta-blocker, an ACE inhibitor, a diuretic, a nitrate, a calcium
  • channel blocker or a statin
  • an agent for treating liver disease such as a corticosteroid, cholestyramine, an interferon, and an anti-viral agent
  • an agent for treating blood disorders such as a corticosteroid, an anti-leukemic agent, or a growth factor
  • an agent for treating immunodeficiency disorders such as gamma globulin.
  • the compounds of the invention can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending 10 upon whether local or systemic treatment is desired and upon the area to be treated.
  • Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • Parenteral administration includes intravenous, intraarterial, subcutaneous,
  • intraperitoneal intramuscular or injection or infusion or intracranial, e.g., intrathecal or
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders.
  • Conventional pharmaceutical 20 carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, 30 suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is
  • the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
  • the compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation 10 types.
  • Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl 15 cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures 20 known in the art.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity 25 of active material calculated to produce the desired therapeutic effect, in association with a
  • the active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, 30 according to the relevant circumstances, including the condition to be treated, the chosen route of
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a 5 homogeneous mixture of a compound of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 10 about 0.1 to about 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which 15 serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present invention can 20 be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar
  • compositions for inhalation or insufflation include solutions and suspensions in
  • compositions 25 pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing 30 device can be attached to a face masks tent, or intermittent positive pressure breathing machine.
  • Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, 5 for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white vaseline, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g., glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and 10 the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its
  • Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization 25 techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical 30 salts.
  • the therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the invention in a pharmaceutical composition 5 can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 g/kg to about 1 g/kg of body weight per day.
  • the dose range is from 10 about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the invention can further include one or more additional ingredients
  • Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating LSD1 in tissue samples, including human, and for identifying LSD1 ligands by inhibition binding of a labeled compound.
  • the present invention includes LSD1 assays that contain such labeled compounds.
  • the present invention further includes isotopically-labeled compounds of the invention.
  • an “isotopically” or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present
  • 3 H also written as T for tritium
  • radio-labeled compounds will depend on the specific application of that radio-labeled compound.
  • a “radio-labeled " or “labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected 5 from the group consisting of 3 H, 14 C, 125 I, 35 S and 82 Br.
  • the compound incorporates 1, 2, or 3 deuterium atoms.
  • the present invention can further include synthetic methods for incorporating radio- isotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily 10 recognize the methods applicable for the compounds of invention.
  • a newly synthesized or identified compound i.e., test compound
  • a test compound which is labeled
  • a test compound can be evaluated for its ability to reduce binding of another compound which is known to bind to LSD1 (i.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to LSD1directly correlates to its binding affinity.
  • the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the 20 labeled standard compound is monitored in order to evaluate the competition between the
  • pH 2 purifications: Waters Sunfire TM C18 5 ⁇ m particle size, 19 x 100 mm column, eluting with mobile phase A: 0.1% TFA (trifluoroacetic acid) in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each 20 compound using the Compound Specific Method Optimization protocol as described in the
  • pH 10 purifications: Waters XBridge C18 5 ⁇ m particle size, 19 x 100 mm column, 25 eluting with mobile phase A: 0.15% NH 4 OH in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [See
  • a reaction vessel containin 6-chloropyridin-2-amine (415 mg, 2.00 mmol), (4-cyanophenyl)boronic acid (353 mg, 2.40 mmol), [1,1'- bis(diphenylphosphino)ferrocene]-dichloropalladium(II) complexed with dichloromethane (1:1) 10 (80 mg, 0.1 mmol) and potassium carbonate (550 mg, 4.0 mmol) in 1,4-dioxane (6 mL) and water (1 mL) was evacuated then refilled with nitrogen. The resulting mixture was heated to 80 oC and stirred for 3 h.
  • NC 2 20 A reaction vessel containin o-2-chloropyridin-3-yl)benzonitrile (320 mg, 1.39 mmol), 4-methyl-8-(4-methylphenyl)-2,6-dioxotetrahydro[1,3,2]oxazaborolo[2,3- b][1,3,2]oxazaborol-4-ium-8-uide (413 mg, 1.67 mmol), [1,1'-
  • Step 2 tert-butyl 3- ⁇ [6-(4-cyanophenyl)-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-8- yl]ethynyl ⁇ pyrrolidine-1-carboxylate
  • a reaction vessel containin o-5-(4- methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile (Example 1, Step 5, 338 mg, 0.868 mmol), 4-methyl-2,6-dioxo-8-vinyltetrahydro[1,3,2]oxazaborolo[2,3-b][1,3,2]oxazaborol-4-ium- 8-uide (206 mg, 1.13 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) 15 complexed with dichloromethane (1:1 ) (42 mg, 0.052 mmol), and potassium carbonate (240 mg, 1.7 mmol) in 1,4-dioxane (6 mL) and water (2 mL) was evacuated then filled with nitrogen.
  • NC 2 10 A reaction vessel containin o-3-chloropyrazin-2-yl)benzonitrile (1.15 g, 5.00 mmol), (4-methylphenyl)boronic acid (0.86 g, 6.4 mmol), sodium carbonate (1.06 g, 10.0 mmol), and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) complexed with dichloromethane (1:1) (0.20 g, 0.25 mmol) in 1,4-dioxane (20.0 mL) and water (4.0 mL) was evacuated then refilled with nitrogen. The resulting mixture was stirred at 110 °C for 3 h then 15 cooled to room temperature.
  • IC 50 data for the example compounds is provided in Table 1 (+ refers to IC50 ⁇ 50 nM; ++ refers to IC50 > 50 nM and ⁇ 100 nM; +++ refers to IC 50 > 50 nM and ⁇ 100 nM; ++++ refers to IC 50 > 500 nM 15 and ⁇ 1000 nM).

Abstract

The present invention is directed to [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5-a]pyrazine derivatives which are LSD1 inhibitors useful in the treatment of diseases such as cancer.

Description

TRIAZOLOPYRIDINES AND TRIAZOLOPYRAZINES AS LSD1 INHIBITORS 5 
FIELD OF THE INVENTION
The present invention is directed to [1,2,4]triazolo[1,5-a]pyridine and [1,2,4]triazolo[1,5- a]pyrazine derivatives which are LSD1 inhibitors useful in the treatment of diseases such as cancer.
10 
BACKGROUND OF THE INVENTION
Epigenetic modifications can impact genetic variation but, when dysregulated, can also contribute to the development of various diseases (Portela, A. and M. Esteller, Epigenetic modifications and human disease. Nat Biotechnol, 2010. 28(10): p. 1057-68; Lund, A.H. and M. 15  van Lohuizen, Epigenetics and cancer. Genes Dev, 2004. 18(19): p. 2315-35). Recently, in depth cancer genomics studies have discovered many epigenetic regulatory genes are often mutated or their own expression is abnormal in a variety of cancers (Dawson, M.A. and T.
Kouzarides, Cancer epigenetics: from mechanism to therapy. Cell, 2012. 150(1): p. 12-27; Waldmann, T. and R. Schneider, Targeting histone modifications--epigenetics in cancer. Curr 20  Opin Cell Biol, 2013. 25(2): p. 184-9; Shen, H. and P.W. Laird, Interplay between the cancer genome and epigenome. Cell, 2013.153(1): p. 38-55). This implies epigenetic regulators function as cancer drivers or are permissive for tumorigenesis or disease progression. Therefore, deregulated epigenetic regulators are attractive therapeutic targets.
One particular enzyme which is associated with human diseases is lysine specific 25  demethylase-1 (LSD1), the first discovered histone demethylase (Shi, Y., et al., Histone
demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell, 2004. 119(7): p. 941-53). It consists of three major domains: the N-terminal SWIRM which functions in nucleosome targeting, the tower domain which is involved in protein-protein interaction, such as transcriptional co-repressor, co-repressor of RE1-silencing transcription factor (CoREST), and 30  lastly the C terminal catalytic domain whose sequence and structure share homology with the flavin adenine dinucleotide (FAD)-dependent monoamine oxidases (i.e., MAO-A and MAO-B)    
(Forneris, F., et al., Structural basis of LSD1-CoREST selectivity in histone H3 recognition. J Biol Chem, 2007. 282(28): p. 20070-4; Anand, R. and R. Marmorstein, Structure and mechanism of lysine-specific demethylase enzymes. J Biol Chem, 2007. 282(49): p. 35425-9; Stavropoulos, P., G. Blobel, and A. Hoelz, Crystal structure and mechanism of human lysine-specific
5  demethylase-1. Nat Struct Mol Biol, 2006. 13(7): p. 626-32; Chen, Y., et al., Crystal structure of human histone lysine-specific demethylase 1 (LSD1). Proc Natl Acad Sci U S A, 2006. 103(38): p. 13956-61). LSD1 also shares a fair degree of homology with another lysine specific demethylase (LSD2) (Karytinos, A., et al., A novel mammalian flavin-dependent histone demethylase. J Biol Chem, 2009. 284(26): p. 17775-82). Although the biochemical mechanism 10  of action is conserved in two isoforms, the substrate specificities are thought to be distinct with relatively small overlap. The enzymatic reactions of LSD1 and LSD2 are dependent on the redox process of FAD and the requirement of a protonated nitrogen in the methylated lysine is thought to limit the activity of LSD1/2 to mono- and di-methylated lysines at the position of 4 or 9 of histone 3 (H3K4 or H3K9). These mechanisms make LSD1/2 distinct from other histone 15  demethylase families (i.e. Jumonji domain containing family) that can demethylate mono-, di-, and tri-methylated lysines through alpha-ketoglutarate dependent reactions (Kooistra, S.M. and K. Helin, Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol, 2012. 13(5): p. 297-311; Mosammaparast, N. and Y. Shi, Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases. Annu Rev 20  Biochem, 2010. 79: p. 155-79).
Methylated histone marks on H3K4 and H3K9 are generally coupled with transcriptional activation and repression, respectively. As part of corepressor complexes (e.g., CoREST), LSD1 has been reported to demethylate H3K4 and repress transcription, whereas LSD1, in nuclear hormone receptor complex (e.g., androgen receptor), may demethylate H3K9 to activate gene 25  expression (Metzger, E., et al., LSD1 demethylates repressive histone marks to promote
androgen-receptor-dependent transcription. Nature, 2005. 437(7057): p. 436-9; Kahl, P., et al., Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res, 2006. 66(23): p. 11341- 7). This suggests the substrate specificity of LSD1 can be determined by associated factors, 30  thereby regulating alternative gene expressions in a context dependent manner. In addition to histone proteins, LSD1 may demethylate non-histone proteins. These include p53 (Huang, J., et    
al., p53 is regulated by the lysine demethylase LSD1. Nature, 2007. 449(7158): p. 105-8.), E2F (Kontaki, H. and I. Talianidis, Lysine methylation regulates E2F1-induced cell death. Mol Cell, 2010. 39(1): p. 152-60), STAT3 (Yang, J., et al., Reversible methylation of promoter-bound STAT3 by histone-modifying enzymes. Proc Natl Acad Sci U S A, 2010. 107(50): p. 21499-504), 5  Tat (Sakane, N., et al., Activation of HIV transcription by the viral Tat protein requires a
demethylation step mediated by lysine-specific demethylase 1 (LSD1/KDM1). PLoS Pathog, 2011. 7(8): p. e1002184), and myosin phosphatase target subunit 1 (MYPT1) (Cho, H.S., et al., Demethylation of RB regulator MYPT1 by histone demethylase LSD1 promotes cell cycle progression in cancer cells. Cancer Res, 2011. 71(3): p. 655-60). The lists of non-histone 10  substrates are growing with technical advances in functional proteomics studies. These suggest additional oncogenic roles of LSD1 beyond regulating chromatin remodeling. LSD1 also associates with other epigenetic regulators, such as DNA methyltransferase 1 (DNMT1) (Wang, J., et al., The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat Genet, 2009. 41(1): p. 125-9) and histone deacetylases (HDACs) complexes 15  (Hakimi, M.A., et al., A core-BRAF35 complex containing histone deacetylase mediates
repression of neuronal-specific genes. Proc Natl Acad Sci U S A, 2002. 99(11): p. 7420-5; Lee, M.G., et al., Functional interplay between histone demethylase and deacetylase enzymes. Mol Cell Biol, 2006. 26(17): p. 6395-402; You, A., et al., CoREST is an integral component of the CoREST- human histone deacetylase complex. Proc Natl Acad Sci U S A, 2001. 98(4): p. 1454- 20  8). These associations augment the activities of DNMT or HDACs. LSD1 inhibitors may
therefore potentiate the effects of HDAC or DNMT inhibitors. Indeed, preclinical studies have shown such potential already (Singh, M.M., et al., Inhibition of LSD1 sensitizes glioblastoma cells to histone deacetylase inhibitors. Neuro Oncol, 2011. 13(8): p. 894-903; Han, H., et al., Synergistic re-activation of epigenetically silenced genes by combinatorial inhibition of DNMTs 25  and LSD1 in cancer cells. PLoS One, 2013. 8(9): p. e75136).
LSD1 has been reported to contribute to a variety of biological processes, including cell proliferation, epithelial-mesenchymal transition (EMT), and stem cell biology (both embryonic stem cells and cancer stem cells) or self-renewal and cellular transformation of somatic cells (Chen, Y., et al., Lysine-specific histone demethylase 1 (LSD1): A potential molecular target for 30  tumor therapy. Crit Rev Eukaryot Gene Expr, 2012. 22(1): p. 53-9; Sun, G., et al., Histone
demethylase LSD1 regulates neural stem cell proliferation. Mol Cell Biol, 2010. 30(8): p. 1997-    
2005; Adamo, A., M.J. Barrero, and J.C. Izpisua Belmonte, LSD1 and pluripotency: a new player in the network. Cell Cycle, 2011. 10(19): p. 3215-6; Adamo, A., et al., LSD1 regulates the balance between self-renewal and differentiation in human embryonic stem cells. Nat Cell Biol, 2011. 13(6): p. 652-9). In particular, cancer stem cells or cancer initiating cells have some 5  pluripotent stem cell properties that contribute to the heterogeneity of cancer cells. This feature may render cancer cells more resistant to conventional therapies, such as chemotherapy or radiotherapy, and then develop recurrence after treatment (Clevers, H., The cancer stem cell: premises, promises and challenges. Nat Med, 2011. 17(3): p. 313-9; Beck, B. and C. Blanpain, Unravelling cancer stem cell potential. Nat Rev Cancer, 2013. 13(10): p. 727-38). LSD1 was 10  reported to maintain an undifferentiated tumor initiating or cancer stem cell phenotype in a
spectrum of cancers (Zhang, X., et al., Pluripotent Stem Cell Protein Sox2 Confers Sensitivity to LSD1 Inhibition in Cancer Cells. Cell Rep, 2013. 5(2): p. 445-57; Wang, J., et al., Novel histone demethylase LSD1 inhibitors selectively target cancer cells with pluripotent stem cell properties. Cancer Res, 2011. 71(23): p. 7238-49). Acute myeloid leukemias (AMLs) are an example of 15  neoplastic cells that retain some of their less differentiated stem cell like phenotype or leukemia stem cell (LSC) potential. Analysis of AML cells including gene expression arrays and chromatin immunoprecipitation with next generation sequencing (ChIP-Seq) revealed that LSD1 may regulate a subset of genes involved in multiple oncogenic programs to maintain LSC (Harris, W.J., et al., The histone demethylase KDM1A sustains the oncogenic potential of MLL- 20  AF9 leukemia stem cells. Cancer Cell, 2012. 21(4): p. 473-87; Schenk, T., et al., Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med, 2012. 18(4): p. 605-11). These findings suggest potential therapeutic benefit of LSD1 inhibitors targeting cancers having stem cell properties, such as AMLs.
25  Overexpression of LSD1 is frequently observed in many types of cancers, including bladder cancer, NSCLC, breast carcinomas, ovary cancer, glioma, colorectal cancer, sarcoma including chondrosarcoma, Ewing’s sarcoma, osteosarcoma, and rhabdomyosarcoma, neuroblastoma, prostate cancer, esophageal squamous cell carcinoma, and papillary thyroid carcinoma. Notably, studies found over-expression of LSD1 was significantly associated with 30  clinically aggressive cancers, for example, recurrent prostate cancer, NSCLC, glioma, breast, colon cancer, ovary cancer, esophageal squamous cell carcinoma, and neuroblastoma. In these    
studies, either knockdown of LSD1 expression or treatment with small molecular inhibitors of LSD1 resulted in decreased cancer cell proliferation and/or induction of apoptosis. See, e.g., Hayami, S., et al., Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int J Cancer, 2011. 128(3): p. 574-86; Lv, T., et al., 5  Over-expression of LSD1 promotes proliferation, migration and invasion in non-small cell lung cancer. PLoS One, 2012. 7(4): p. e35065; Serce, N., et al., Elevated expression of LSD1 (Lysine- specific demethylase 1) during tumour progression from pre-invasive to invasive ductal carcinoma of the breast. BMC Clin Pathol, 2012. 12: p. 13; Lim, S., et al., Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker 10  predicting aggressive biology. Carcinogenesis, 2010. 31(3): p. 512-20; Konovalov, S. and I.
Garcia-Bassets, Analysis of the levels of lysine-specific demethylase 1 (LSD1) mRNA in human ovarian tumors and the effects of chemical LSD1 inhibitors in ovarian cancer cell lines. J Ovarian Res, 2013. 6(1): p. 75; Sareddy, G.R., et al., KDM1 is a novel therapeutic target for the treatment of gliomas. Oncotarget, 2013. 4(1): p. 18-28; Ding, J., et al., LSD1-mediated epigenetic 15  modification contributes to proliferation and metastasis of colon cancer. Br J Cancer, 2013.
109(4): p. 994-1003; Bennani-Baiti, I.M., et al., Lysine-specific demethylase 1
(LSD1/KDM1A/AOF2/BHC110) is expressed and is an epigenetic drug target in
chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma. Hum Pathol, 2012. 43(8): p. 1300-7; Schulte, J.H., et al., Lysine-specific demethylase 1 is strongly expressed in20  poorly differentiated neuroblastoma: implications for therapy. Cancer Res, 2009. 69(5): p. 2065- 71; Crea, F., et al., The emerging role of histone lysine demethylases in prostate cancer. Mol Cancer, 2012. 11: p. 52; Suikki, H.E., et al., Genetic alterations and changes in expression of histone demethylases in prostate cancer. Prostate, 2010. 70(8): p. 889-98; Yu, Y., et al., High expression of lysine-specific demethylase 1 correlates with poor prognosis of patients with 25  esophageal squamous cell carcinoma. Biochem Biophys Res Commun, 2013. 437(2): p. 192-8;
Kong, L., et al., Immunohistochemical expression of RBP2 and LSD1 in papillary thyroid carcinoma. Rom J Morphol Embryol, 2013. 54(3): p. 499-503.
Recently, the induction of CD86 expression by inhibiting LSD1 activity was reported (Lynch, J.T., et al., CD86 expression as a surrogate cellular biomarker for pharmacological 30  inhibition of the histone demethylase lysine-specific demethylase 1. Anal Biochem, 2013. 442(1): p. 104-6). CD86 expression is a marker of maturation of dendritic cells (DCs) which are    
involved in antitumor immune response. Notably, CD86 functions as a co-stimulatory factor to activate T cell proliferation (Greaves, P. and J.G. Gribben, The role of B7 family molecules in hematologic malignancy. Blood, 2013. 121(5): p. 734-44; Chen, L. and D.B. Flies, Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol, 2013. 13(4): p. 227- 5  42).
In addition to playing a role in cancer, LSD1 activity has also been associated with viral pathogenesis. Particularly, LSD1 activity appears to be linked with viral replications and expressions of viral genes. For example, LSD1 functions as a co-activator to induce gene expression from the viral immediate early genes of various type of herpes virus including herpes 10  simplex virus (HSV), varicella zoster virus (VZV), and -herpesvirus human cytomegalovirus (Liang, Y., et al., Targeting the JMJD2 histone demethylases to epigenetically control herpesvirus infection and reactivation from latency. Sci Transl Med, 2013. 5(167): p. 167ra5; Liang, Y., et al., Inhibition of the histone demethylase LSD1 blocks alpha-herpesvirus lytic replication and reactivation from latency. Nat Med, 2009. 15(11): p. 1312-7). In this setting, a 15  LSD1 inhibitor showed antiviral activity by blocking viral replication and altering virus
associated gene expression.
Recent studies have also shown that the inhibition of LSD1 by either genetic depletion or pharmacological intervention increased fetal globin gene expression in erythroid cells (Shi, L., et al., Lysine-specific demethylase 1 is a therapeutic target for fetal hemoglobin induction. Nat 20  Med, 2013. 19(3): p.291-4; Xu, J., et al., Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc Natl Acad Sci U S A, 2013. 110(16): p. 6518-23). Inducing fetal globin gene would be potentially therapeutically beneficial for the disease of -globinopathies, including -thalassemia and sickle cell disease where the production of normal β-globin, a component of adult hemoglobin, is impaired (Sankaran, V.G. and S.H. Orkin, The switch from 25  fetal to adult hemoglobin. Cold Spring Harb Perspect Med, 2013. 3(1): p. a011643; Bauer, D.E., S.C. Kamran, and S.H. Orkin, Reawakening fetal hemoglobin: prospects for new therapies for the beta-globin disorders. Blood, 2012. 120(15): p. 2945-53). Moreover, LSD1 inhibition may potentiate other clinically used therapies, such as hydroxyurea or azacitidine. These agents may act, at least in part, by increasing -globin gene expression through different mechanisms.
30  In summary, LSD1 contributes to tumor development by altering epigenetic marks on histones and non-histone proteins. Accumulating data have validated that either genetic    
depletion or pharmacological intervention of LSD1 normalizes altered gene expressions, thereby inducing differentiation programs into mature cell types, decreasing cell proliferation, and promoting apoptosis in cancer cells. Therefore, LSD1 inhibitors alone or in combination with established therapeutic drugs would be effective to treat the diseases associated with LSD1 5  activity. SUMMARY OF THE INVENTION
The present invention is directed to, inter alia, a compound of Formula I:
 
 
Figure imgf000008_0001
tituent variables are defined herein.
The present invention is further directed to a pharmaceutical composition comprising a compound of Formula I and at least one pharmaceutically acceptable carrier.
The present invention is further directed to a method of inhibiting LSD1 comprising 15  contacting the LSD1 with a compound of Formula I.
The present invention is further directed to a method of treating an LSD1-mediated disease in a patient comprising administering to the patient a therapeutically effective amount of a compound of Formula I. 20  DETAILED DESCRIPTION
The present invention provides, inter alia, LSD1-inhibiting compounds such as a compound of Formula I:
   
 
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein:
X is N or CRX;
5  Ring A is C6-10 aryl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA;
Ring B is C6-10 aryl; 5-10 membered heteroaryl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; C3-10 cycloalkyl; or 4-10 membered heterocycloalkyl 10  comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S; wherein said C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
15  NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, or
S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
20  NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and
S(O)2NRc1Rd1;
wherein when X is CRX, then R1 is not CN;
R2 is H, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy2, CN, ORa2, SRa2, 25  C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2,
NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2,    
NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, or
S(O)2NRc2Rd2; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy2, halo, CN, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, 5  NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and
S(O)2NRc2Rd2;
each RA is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, OC(O)NRc4Rd4, 10  NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4C(O)NRc4Rd4, C(=NRe4)Rb4, C(=NRe4)NRc4Rd4, NRc4C(=NRe4)NRc4Rd4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4,
S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3, substituents independently selected from halo, C1-6 haloalkyl, CN, NO2, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, 15  OC(O)NRc4Rd4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4C(O)NRc4Rd4, C(=NRe4)Rb4, C(=NRe4)NRc4Rd4, NRc4C(=NRe4)NRc4Rd4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4;
each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5,
20  OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(=NRe5)Rb5, C(=NRe5)NRc5Rd5, NRc5C(=NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3 substituents independently selected from Cy3, halo, C1-6 haloalkyl, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, 25  OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(=NRe5)Rb5, C(=NRe5)NRc5Rd5, NRc5C(=NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5;
RX is independently selected from H, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, 30  NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7C(O)NRc7Rd7, C(=NRe7)Rb7, C(=NRe7)NRc7Rd7,
   
NRc7C(=NRe7)NRc7Rd7, NRc7S(O)Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, S(O)Rb7,
S(O)NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7;
each Cy1, Cy2, Cy3, and Cy4 is independently selected from C6-10 aryl, C3-10 cycloalkyl, 5- 10 membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally 5  substituted with 1, 2, 3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, CN, NO2, ORa6, SRa6, C(O)Rb6, 10  C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6,
NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6,
NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and
S(O)2NRc6Rd6, wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 15  alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted by 1, 2, or 3 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, 20  S(O)NRc6Rd6, S(O) 6
2Rb6, and S(O)2NRc Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and Cy4; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3,
25  NRc3C(O)NRc3Rd3, C(=NRe3)Rb3, C(=NRe3)NRc3Rd3, NRc3C(=NRe3)NRc3Rd3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3;
each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 30  C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6
alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered    
heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, 5  NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 10  aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally 15  substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
20  each Ra2, Rb2, Rc2, and Rd2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25  heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, 30  S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
   
or any Rc2 and Rd2 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7,
5  C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, 10  C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, 15  C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 20  C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
25  or any Rc3 and Rd3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
30  NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O) b7
2R , NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4-    
7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
5  NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra4, Rb4, Rc4, and Rd4 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 10  4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc4 and Rd4 together with the N atom to which they are attached form a 4-, 5-, 6-, 15  or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
20  each Ra5, Rb5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25  heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, 30  S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O) c7
2NR Rd7;
   
or any Rc5 and Rd5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, 5  C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, 10  C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, 15  C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, 20  NRc7S(O) c7
2NR Rd7, and S(O) 7
2NRc Rd7;
or any Rc6 and Rd6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
25  NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7,
S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra7, Rb7, Rc7, and Rd7 is independently selected from H, C1-4 alkyl, C1-4 haloalkyl, C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, 30  halo, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, C1-4 alkylamino, di(C1-4 alkyl)amino, C1-4 haloalkyl, and C1-4 haloalkoxy; and    
each Re1, Re2, Re3, Re4, Re5, Re6, and Re7 is independently selected from H, C1-4 alkyl, and CN. In some embodiments:
5  X is N or CRX;
Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA;
Ring B is phenyl or 5-6 membered heteroaryl comprising carbon and 1, 2, 3 or 4
10  heteroatoms selected from N, O, and S; wherein said phenyl and 5-6 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, 15  NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O) b1
2R , or
S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, 20  NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and
S(O)2NRc1Rd1;
wherein when X is CRX, then R1 is not CN;
R2 is H, halo, C1-6 alkyl, CN, ORa2, C(O)Rb2, C(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, S(O)2Rb2, or S(O)2NRc2Rd2; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 25  substituents independently selected from halo, CN, ORa2, C(O)Rb2, C(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, S(O)2Rb2, and S(O)2NRc2Rd2;
each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected 30  from halo, C d4
1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4R , C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4;    
each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 5
1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5, NRc C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1 , 2, or 3 substituents independently selected from Cy3, halo, C1-6 5  haloalkyl, CN, NO a5
2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)OR , NRc5Rd5, NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5;
RX is independently selected from H, halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, NRc7Rd7, NRc7C(O)Rb7, S(O)2Rb7, and S(O)2NRc7Rd7;
each Cy1, Cy3, and Cy4 is independently selected from C6-10 aryl, C3-10 cycloalkyl, 5-10 10  membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally
substituted with 1, 2, 3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 15  alkyl-, and (4-7 membered heterocycloalkyl)-C a6
1-4 alkyl-, CN, NO2, ORa6, SR , C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6,
NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6,
NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and
S(O)2NRc6Rd6, wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 20  membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted by 1, 2, or 3 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, 25  NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, and Cy4; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3; 30  each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered    
heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 5  C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
10  or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
15  NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O) b7
2R , NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, 20  NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra2, Rb2, Rc2, and Rd2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered25  heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 30  1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,    
NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O) c7
2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NR Rd7;
or any Rc2 and Rd2 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
5  independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 10  7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally
substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, 15  and S(O) 7
2NRc Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 20  alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, 25  NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc3 and Rd3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 30  5-6 membered heteroaryl, C 7
1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,    
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, 5  C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O) 7
2NRc7Rd ;
each Ra4, Rb4, Rc4, and Rd4 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, 10  C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, 15  NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc4 and Rd4 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
20  NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7,
S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra5, Rb5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- 25  C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6
alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, 30  halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,
   
NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc5 and Rd5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
5  independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 10  7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally
substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, 15  and S(O)2NRc7Rd7;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
20  OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc6 and Rd6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents
25  independently selected from C a7
1-6 alkyl, C1-6 haloalkyl, halo, CN, OR , SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra7, Rb7, Rc7, and Rd7 is independently selected from H, C1-4 alkyl, C1-4 haloalkyl, 30  C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino,    
halo, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, C1-4 alkylamino, di(C1-4 alkyl)amino, C1-4 haloalkyl, and C1-4 haloalkoxy; and
each Re1, Re6, and Re7 is independently selected from H, C1-4 alkyl, and CN. 5  In some embodiments:
X is N or CRX;
Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from RA;
Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected 10  from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, S(O)2Rb1, or S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, C(O)Rb1, C(O)NRc1Rd1, 15  C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, S(O)2Rb1, and S(O) d1
2NRc1R ;
R2 is H;
each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, and ORa4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from CN and ORa4;
20  each RB is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, and ORa5;
RX is H;
each Cy1 and Cy4 is independently selected from phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 25  3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6, 30  wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, are each optionally substituted by 1, 2, or 3 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN,    
NO 6 d6 2, ORa , SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, NRc6R , NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6,
NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, and 4-7 membered
5  heterocycloalkyl; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3, and wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, 10  NRc6C(O)Rb6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2- 15  6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,
20  NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7,
NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 25  aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents 30  independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,    
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7,
NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H and C1-6 alkyl; each Ra4 is independently selected from H and C1-6 alkyl;
5  each Ra5 is independently selected from H and C1-6 alkyl;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H and C1-6 alkyl; and each Ra7, Rb7, Rc7, and Rd7 is independently selected from H and C1-4 alkyl. In some embodiments, X is N.
10  In some embodiments, X is CRX.
In some embodiments, Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA.
15  In some embodiments, Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from RA.
In some embodiments, Ring A is phenyl substituted by one RA.
In some embodiments, Ring A is phenyl substituted by CN.
In some embodiments, Ring B is phenyl or 5-6 membered heteroaryl comprising carbon 20  and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; wherein said phenyl and 5-6 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB.
In some embodiments, Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected from RB.
25  In some embodiments, Ring B is phenyl substituted by one RB.
In some embodiments, Ring B is phenyl substituted by methyl.
In some embodiments, R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, S(O)2Rb1, or S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each 30  optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, S(O)2Rb1, and S(O)2NRc1Rd1.    
In some embodiments, R1 is C1-6 alkyl, Cy1, or ORa1, wherein said C1-6 alkyl is substituted with one Cy1.
In some embodiments, R1 is pyrrolidin-3-ylmethoxy, 2-pyrrolidin-3-ylethyl, (1- methylpyrrolidin-3-yl)ethyl, 3-[(methylamino)methyl]phenyl, 3-aminopyrrolidin-1- 5  yl)methyl]phenyl, piperazin-1-ylmethyl, 4-methylpiperazin-1-yl)methyl, 3- (dimethylamino)pyrrolidin-1-yl, 3-(methylamino)pyrrolidin-1-yl, or (1-methylpyrrolidin-3- yl)methoxy.
In some embodiments, R2 is H.
In some embodiments, each RA is independently selected from halo, C1-6 alkyl, C1-6 10  haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O) b4
2R , and S(O)2NRc4Rd4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4.
In some embodiments, each RA is independently selected from halo, C1-6 alkyl, C1-6 15  haloalkyl, CN, and ORa4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3,
substituents independently selected from CN and ORa4.
In some embodiments, RA is CN.
In some embodiments, each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, 20  NRc5Rd5, NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3 substituents independently selected from Cy3, halo, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5, NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5.
In some embodiments, each RB is independently selected from halo, C1-6 alkyl, C2-6 25  alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, and ORa5.
In some embodiments, RB is C1-6 alkyl.
In some embodiments, RB is methyl.
In some embodiments, RX is H.
In some embodiments, each Cy1 is independently selected from phenyl, C3-7 cycloalkyl, 30  5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy.    
In some embodiments, each Cy1 is phenyl or 4-7 membered heterocycloalkyl, each optionally substituted with 1 or 2 substituents independently selected from RCy.
In some embodiments, each Cy1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from RCy.
5  In some embodiments, each Cy1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from C1-4 alkyl and NRc6Rd6, wherein said C1-4 alkyl is optionally substituted with NRc6Rd6.
In some embodiments, each RCy is C1-4 alkyl and NRc6Rd6, wherein said C1-4 alkyl is optionally substituted with NRc6Rd6.
10  In some embodiments, each Ra1 is independently selected from H, C1-6 alkyl, and Cy4; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3.
In some embodiments, each Ra1 is independently selected from H, C1-6 alkyl, and 4-7 15  membered heterocycloalkyl; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3
substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3,
C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3, and wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, 20  C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, S(O)2Rb6, and S(O) d6
2NRc6R .
In some embodiments, each Ra1 is C1-4 alkyl substituted by 4-7 membered
heterocycloalkyl, wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, S(O)2Rb6, and
25  S(O) 6
2NRc Rd6.
In some embodiments, each Ra1 is pyrrolidinylmethyl optionally substituted with one C1-4 alkyl.
In some embodiments, the compounds of the invention have Formula IIa:
   
Figure imgf000027_0001
In some embodiments, the compounds of the invention have Formula IIb: 5 
Figure imgf000027_0002
In some embodiments, the compounds of the invention have Formula IIIa:
Figure imgf000027_0003
 
In some embodiments, the compounds of the invention have Formula IIIb:    
Figure imgf000028_0001
It is appreciated that certain features of the invention, which are, for clarity, described in 5  the context of separate embodiments, can also be provided in combination in a single
embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable
subcombination.
As used herein, the phrase "optionally substituted" means unsubstituted or substituted. As 10  used herein, the term "substituted" means that a hydrogen atom is removed and replaced by a monovalent substituent, or two hydrogen atoms are replaced with a divalent substituent like a terminal oxo group. It is to be understood that substitution at a given atom is limited by valency.
Throughout the definitions, the term "Ci-j" indicates a range which includes the endpoints, wherein i and j are integers and indicate the number of carbons. Examples include C1-4, C1-6, and 15  the like.
The term "z-membered" (where z is an integer) typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is z. For example, piperidinyl is an example of a 6-membered heterocycloalkyl ring, pyrazolyl is an example of a 5- membered heteroaryl ring, pyridyl is an example of a 6-membered heteroaryl ring, and 1, 2, 3, 4- 20  tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
As used herein, the term "Ci-j alkyl," employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched, having i to j carbons. In some embodiments, the alkyl group contains from 1 to 6 carbon atoms or from 1 to 4 carbon atoms, or from 1 to 3 carbon atoms. Examples of alkyl moieties include, but are not25  limited to, chemical groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, s-butyl, and t- butyl.    
As used herein, the term "Ci-j alkoxy," employed alone or in combination with other terms, refers to a group of formula -O-alkyl, wherein the alkyl group has i to j carbons. Example alkoxy groups include methoxy, ethoxy, and propoxy (e.g., n-propoxy and isopropoxy). In some embodiments, the alkyl group has 1 to 3 carbon atoms.
5  As used herein, "Ci-j alkenyl," employed alone or in combination with other terms, refers to an unsaturated hydrocarbon group having one or more double carbon-carbon bonds and having i to j carbons. In some embodiments, the alkenyl moiety contains 2 to 6 or 2 to 4 carbon atoms. Example alkenyl groups include, but are not limited to, ethenyl, n-propenyl, isopropenyl, n-butenyl, sec-butenyl, and the like.
10  As used herein, "Ci-j alkynyl," employed alone or in combination with other terms, refers to an unsaturated hydrocarbon group having one or more triple carbon-carbon bonds and having i to j carbons. Example alkynyl groups include, but are not limited to, ethynyl, propyn-1-yl, propyn-2-yl, and the like. In some embodiments, the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
15  As used herein, the term "Ci-j alkylamino," employed alone or in combination with other terms, refers to a group of formula -NH(alkyl), wherein the alkyl group has i to j carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.  In some embodiments, the alkylamino group is–NH(C1-4 alkyl) such as, for example, methylamino, ethylamino or propylamino.
20  As used herein, the term "di-Ci-j-alkylamino," employed alone or in combination with other terms, refers to a group of formula -N(alkyl)2, wherein each of the two alkyl groups has, independently, i to j carbon atoms. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms. In some embodiments, the dialkylamino group is–N(C1-4 alkyl)2 such as, for example, dimethylamino or diethylamino.
25  As used herein, the term "Ci-j alkylthio," employed alone or in combination with other terms, refers to a group of formula -S-alkyl, wherein the alkyl group has i to j carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. In some embodiments, the alkylthio group is C1-4 alkylthio such as, for example, methylthio or ethylthio.
As used herein, the term "amino," employed alone or in combination with other terms, 30  refers to a group of formula–NH2.
   
As used herein, the term "aryl," employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic hydrocarbon, such as, but not limited to, phenyl, 1-naphthyl, 2-naphthyl, anthracenyl, phenanthrenyl, and the like. In some embodiments, aryl is C6-10 aryl. In some embodiments, the aryl group is a
5  naphthalene ring or phenyl ring. In some embodiments, the aryl group is phenyl.
As used herein, the term "aryl-Ci-j alkyl," employed alone or in combination with other terms, refers to an alkyl group substituted by an aryl group. An example of a aryl-Ci-j alkyl group is benzyl.
As used herein, the term "carbonyl", employed alone or in combination with other terms, 10  refers to a -C(O)- group.
As used herein, the term "Ci-j cycloalkyl," employed alone or in combination with other terms, refers to a non-aromatic cyclic hydrocarbon moiety having i to j ring-forming carbon atoms, which may optionally contain one or more alkenylene groups as part of the ring structure. Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) ring 15  systems. Also included in the definition of cycloalkyl are moieties that have one or more
aromatic rings fused (i.e.,, having a bond in common with) to the cycloalkyl ring, for example, benzo derivatives of cyclopentane, cyclopentene, cyclohexane, and the like. One or more ring- forming carbon atoms of a cycloalkyl group can be oxidized to form carbonyl linkages. In some embodiments, cycloalkyl is C3-10 cycloalkyl, C3-7 cycloalkyl, or C5-6 cycloalkyl. Exemplary 20  cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl,
cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, and the like. Further exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
As used herein, the term "Ci-j cycloalkyl-Ci-j alkyl," employed alone or in combination 25  with other terms, refers to an alkyl group substituted by a cycloalkyl group. An example of a Ci-j cycloalkyl-Ci-j alkyl group is cyclopropylmethyl.
As used herein, "Ci-j haloalkoxy," employed alone or in combination with other terms, refers to a group of formula–O-haloalkyl having i to j carbon atoms. An example haloalkoxy group is OCF3. An additional example haloalkoxy group is OCHF2. In some embodiments, the 30  haloalkoxy group is fluorinated only. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. In some embodiments, the haloalkoxy group is C1-4 haloalkoxy.    
As used herein, the term "halo," employed alone or in combination with other terms, refers to a halogen atom selected from F, Cl, I or Br. In some embodiments, "halo" refers to a halogen atom selected from F, Cl, or Br. In some embodiments, the halo substituent is F.
As used herein, the term "Ci-j haloalkyl," employed alone or in combination with other 5  terms, refers to an alkyl group having from one halogen atom to 2s+1 halogen atoms which may be the same or different, where "s" is the number of carbon atoms in the alkyl group, wherein the alkyl group has i to j carbon atoms. In some embodiments, the haloalkyl group is fluorinated only. In some embodiments, the haloalkyl group is fluoromethyl, difluoromethyl, or
trifluoromethyl. In some embodiments, the haloalkyl group is trifluoromethyl. In some
10  embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "heteroaryl," employed alone or in combination with other terms, refers to a monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings) aromatic heterocylic moiety, having one or more heteroatom ring members selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl group has 1, 2, 3, or 4 heteroatom ring 15  members. In some embodiments, the heteroaryl group has 1, 2, or 3 heteroatom ring members.
In some embodiments, the heteroaryl group has 1 or 2 heteroatom ring members. In some embodiments, the heteroaryl group has 1 heteroatom ring member. In some embodiments, the heteroaryl group is 5- to 10-membered or 5- to 6-membered. In some embodiments, the heteroaryl group is 5-membered. In some embodiments, the heteroaryl group is 6-membered. 20  When the heteroaryl group contains more than one heteroatom ring member, the heteroatoms may be the same or different. The nitrogen atoms in the ring(s) of the heteroaryl group can be oxidized to form N-oxides. Example heteroaryl groups include, but are not limited to, pyridine, pyrimidine, pyrazine, pyridazine, pyrrole, pyrazole, azolyl, oxazole, isoxazole, thiazole, isothiazole, imidazole, furan, thiophene, triazole, tetrazole, thiadiazole, quinoline, isoquinoline, 25  indole, benzothiophene, benzofuran, benzisoxazole, imidazo[1, 2-b]thiazole, purine, triazine, and the like.
A 5-membered heteroaryl is a heteroaryl group having five ring-forming atoms comprising wherein one or more of the ring-forming atoms are independently selected from N, O, and S. In some embodiments, the 5-membered heteroaryl group has 1, 2, or 3 heteroatom ring 30  members. In some embodiments, the 5-membered heteroaryl group has 1 or 2 heteroatom ring members. In some embodiments, the 5-membered heteroaryl group has 1 heteroatom ring    
member. Example ring-forming members include CH, N, NH, O, and S. Example five- membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1, 2, 3-triazolyl, tetrazolyl, 1, 2, 3-thiadiazolyl, 1, 2, 3-oxadiazolyl, 1, 2, 4-triazolyl, 1, 2, 4-thiadiazolyl, 1, 2, 4-oxadiazolyl, 1, 3, 4-triazolyl, 1, 3, 4-thiadiazolyl, and 1, 3, 5  4-oxadiazolyl.
A 6-membered heteroaryl is a heteroaryl group having six ring-forming atoms wherein one or more of the ring-forming atoms is N. In some embodiments, the 6-membered heteroaryl group has 1, 2, or 3 heteroatom ring members. In some embodiments, the 6-membered heteroaryl group has 1 or 2 heteroatom ring members. In some embodiments, the 6-membered 10  heteroaryl group has 1 heteroatom ring member. Example ring-forming members include CH and N. Example six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl, and pyridazinyl.
As used herein, the term "heteroaryl-Ci-j alkyl," employed alone or in combination with other terms, refers to an alkyl group substituted by a heteroaryl group. An example of a
15  heteroaryl-Ci-j alkyl group is pyridylmethyl.
As used herein, the term "heterocycloalkyl," employed alone or in combination with other terms, refers to non-aromatic heterocyclic ring system, which may optionally contain one or more unsaturations as part of the ring structure, and which has at least one heteroatom ring member independently selected from nitrogen, sulfur and oxygen. In some embodiments, the 20  heterocycloalkyl group has 1, 2, 3, or 4 heteroatom ring members. In some embodiments, the heterocycloalkyl group has 1, 2, or 3 heteroatom ring members. In some embodiments, the heterocycloalkyl group has 1 or 2 heteroatom ring members. In some embodiments, the heterocycloalkyl group has 1 heteroatom ring member. When the heterocycloalkyl group contains more than one heteroatom in the ring, the heteroatoms may be the same or different. 25  Example ring-forming members include CH, CH2, C(O), N, NH, O, S, S(O), and S(O)2.
Heterocycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) ring systems, including spiro systems. Also included in the definition of heterocycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the non- aromatic ring, for example, 1, 2, 3, 4-tetrahydro-quinoline, dihydrobenzofuran and the like. The 30  carbon atoms or heteroatoms in the ring(s) of the heterocycloalkyl group can be oxidized to form a carbonyl, sulfinyl, or sulfonyl group (or other oxidized linkage) or a nitrogen atom can be    
quaternized. In some embodiments, the heterocycloalkyl is 5- to 10-membered, 4- to 10- membered, 4- to 7-membered, 5-membered, or 6-membered. Examples of heterocycloalkyl groups include 1, 2, 3, 4-tetrahydro-quinoline, dihydrobenzofuran, azetidine, azepane, pyrrolidine, piperidine, piperazine, morpholine, thiomorpholine, and pyran.
5  As used herein, the term "heterocycloalkyl-Ci-j alkyl," employed alone or in combination with other terms, refers to an alkyl group substituted by a heterocycloalkyl group. An example of a heterocycloalkyl-Ci-j alkyl group is pyrrolidinylmethyl.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereoisomers, are intended unless 10  otherwise indicated. Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, 15  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.
When the compounds of the invention contain a chiral center, the compounds can be any of the possible stereoisomers. In compounds with a single chiral center, the stereochemistry of 20  the chiral center can be (R) or (S). In compounds with two chiral centers, the stereochemistry of the chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R) and (R), (R) and (S); (S) and (R), or (S) and (S). In compounds with three chiral centers, the stereochemistry each of the three chiral centers can each be independently (R) or (S) so the configuration of the chiral centers can be (R), (R) and (R); (R), (R) and (S); (R), (S) and 25  (R); (R), (S) and (S); (S), (R) and (R); (S), (R) and (S); (S), (S) and (R); or (S), (S) and (S).
Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallization using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and 30  L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as -camphorsulfonic acid.    
Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of -methylbenzylamine (e.g., S and R forms, or diastereoisomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1, 2- diaminocyclohexane, and the like.
5  Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Compounds of the invention also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant 10  migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric
protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone– enol pairs, amide - imidic acid pairs, lactam– lactim pairs, amide - imidic acid pairs, enamine– imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 15  1, 2, 4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers.
20  The term "compound" as used herein is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified.
All compounds, and pharmaceutically acceptable salts thereof, can be found together 25  with other substances such as water and solvents (e.g., hydrates and solvates) or can be isolated.
In some embodiments, the compounds of the invention, or salts thereof, are substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in a compound of the invention. 30  Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least    
about 97%, or at least about 99% by weight of the compounds of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound 5  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.
The expressions, "ambient temperature" and "room temperature," as used herein, are understood in the art, and refer generally to a temperature, e.g., a reaction temperature, that is 10  about the temperature of the room in which the reaction is carried out, for example, a
temperature from about 20 ºC to about 30 ºC.
The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or 15  base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of 20  the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, 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, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or 25  butanol) or acetonitrile (MeCN) are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th Ed., (Mack Publishing Company, Easton, 1985), p. 1418, Berge et al., J. Pharm. Sci., 1977, 66(1), 1-19, and in Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, (Wiley, 2002).
The following abbreviations may be used herein: AcOH (acetic acid); Ac2O (acetic30  anhydride); aq. (aqueous); atm. (atmosphere(s)); Boc (t-butoxycarbonyl); BOP ((benzotriazol-1- yloxy)tris(dimethylamino)phosphonium hexafluorophosphate); br (broad); Cbz (carboxybenzyl);    
calc. (calculated); d (doublet); dd (doublet of doublets); DBU (1,8-diazabicyclo[5.4.0]undec-7- ene); DCM (dichloromethane); DIAD (N, N'-diisopropyl azidodicarboxylate); DIEA (N,N- diisopropylethylamine); DIPEA (N, N-diisopropylethylamine); DMF (N, N-dimethylformamide); Et (ethyl); EtOAc (ethyl acetate); g (gram(s)); h (hour(s)); HATU (N, N, N', N'-tetramethyl-O-(7- 5  azabenzotriazol-1-yl)uronium hexafluorophosphate); HCl (hydrochloric acid); HPLC (high
performance liquid chromatography); Hz (hertz); IPA (isopropyl alcohol); J (coupling constant); LCMS (liquid chromatography– mass spectrometry); m (multiplet); M (molar); mCPBA (3- chloroperoxybenzoic acid); MS (Mass spectrometry); Me (methyl); MeCN (acetonitrile); MeOH (methanol); mg (milligram(s)); min. (minutes(s)); mL (milliliter(s)); mmol (millimole(s)); N 10  (normal); nM (nanomolar); NMP (N-methylpyrrolidinone); NMR (nuclear magnetic resonance spectroscopy); OTf (trifluoromethanesulfonate); Ph (phenyl); pM (picomolar); RP-HPLC (reverse phase high performance liquid chromatography); s (singlet); t (triplet or tertiary); TBS (tert-butyldimethylsilyl); tert (tertiary); tt (triplet of triplets); TFA (trifluoroacetic acid); THF (tetrahydrofuran); µg (microgram(s)); µL (microliter(s)); µM (micromolar); wt % (weight 15  percent). Synthesis
Compounds of the invention, including salts thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible 20  synthetic routes.
The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the
intermediates, or products at the temperatures at which the reactions are carried out, e.g., 25  temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Preparation of compounds of the invention can involve the protection and deprotection of 30  various chemical groups. The need for protection and deprotection, and the selection of
appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry    
of protecting groups can be found, for example, in P. G. M. Wuts and T. W. Greene, Protective Groups in Organic Synthesis, 4th Ed., Wiley & Sons, Inc., New York (2006), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For 5  example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV- visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectroscopy (LCMS), or thin layer chromatography (TLC). Compounds can be purified by those skilled in the art by a variety of 10  methods, including high performance liquid chromatography (HPLC) ("Preparative LC-MS Purification: Improved Compound Specific Method Optimization" Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs J. Combi. Chem. 2004, 6(6), 874-883, which is incorporated herein by reference in its entirety) and normal phase silica chromatography.
Compounds of formula 9 can be synthesized as shown in Scheme 1. Compound 1 can 15  undergo Suzuki reaction with an appropriate boronic acid or ester of formula 2 in the presence of a palladium catalyst and a suitable base such as K2CO3 to provide compound of formula 3.
Installation of ring B to give compound of formula 5 can be achieved by coupling of compound 3 with compound of formula 4 under standard Suzuki coupling conditions (M is a boronic acid or ester, with palladium catalysis), or standard Negishi coupling conditions (M is Zn-halo, in the 20  presence of a palladium catalyst), or standard Buchwald amination conditions (M is H attached to a ring-forming N atom in ring B, in the presence of a palladium catalyst and a suitable base). Halogenation of compound 5 using N-chlorosuccinimide, N-bromosuccinimide or N- iodosuccinimide can provide a compound of formula 6 (Hal is Cl, Br or I). Compound 6 can be converted to a formamidoxime derivative of formula 7 by reacting with N,N-dimethylformamide 25  dimethyl acetal, followed by treatment with hydroxylamine. The formamidoxime derivative 7 can undergo cyclization upon treating with trifluoroacetic anhydride (TFAA) to afford a triazole compound of formula 8. Finally, the aryl halide 8 can react with R1-M to give a compound of formula 9 under standard cross coupling conditions, such as Suzuki coupling conditions (M is a boronic acid or ester, with palladium catalysis), Sonogashira coupling conditions (M is a
30  terminal alkynyl, with palladium catalysis), Negishi coupling conditions (M is ZnCl, ZnBr or ZnI, with palladium catalysis), Buchwald amination conditions (R1-M is an amine (M is H), with    
palladium catalysis) or Ullmann coupling conditions (R1-M is an alcohol (M is H), with palladium or copper catalysis).
Scheme 1
2 M
l 2
Figure imgf000038_0001
MeO
NMe2 A A
X Hal A
X Hal X R1
1-
  5 
Figure imgf000038_0002
, - , ized as shown in Scheme 2. Compound 6, which can be prepared as described in Scheme 1, can react with a nitrile R2-CN to deliver a triazole compound of formula 10 via a copper-catalyzed tandem addition−oxidative cyclization. This tandem reaction is described in Nagasawa et. al. in J. Am. Chem. Soc. 2009, 131, 42, 15080. Finally, the aryl halide 10 can react with R1-M under standard 10  cross coupling conditions as described in Scheme 1 (e.g., Suzuki coupling, Negishi coupling, Sonogashira coupling, Buchwald amination or Ullmann coupling) to give compounds of formula 11.
Scheme 2 1 2  
Figure imgf000038_0003
15  Alternatively, compounds of formula 11 can be prepared as shown in Scheme 3. Aryl halide 6 can react with R1-M under standard cross coupling conditions as described in Scheme 1 (e.g., Suzuki coupling, Negishi coupling, Sonogashira coupling, Buchwald amination or Ullmann coupling) to give compounds of formula 12. Condensation of amino-pyridine derivative 12 with ethoxycarbonyl isothiocyanate, followed by treatment with hydroxylamine can give the    
aminotriazole of formula 13. Transformation of the amino group in compound 13 to bromide can be achieved under standard Sandmeyer reaction conditions (e.g., NaNO2, HBr then CuBr) to give compounds of formula 14. Functionalization of aryl bromide 14 with an R2 substituent to give compound 11 can be performed under standard cross coupling reaction conditions (e.g., Suzuki 5  coupling, Negishi coupling, Sonogashira coupling, Buchwald amination or Ullmann coupling) as described in the previous Schemes.
Scheme 3
2
Figure imgf000039_0001
10  Methods of Use
Compounds of the invention are LSD1 inhibitors and, thus, are useful in treating diseases and disorders associated with activity of LSD1. For the uses described herein, any of the compounds of the invention, including any of the embodiments thereof, may be used.
In some embodiments, the compounds of the invention are selective for LSD1 over 15  LSD2, meaning that the compounds bind to or inhibit LSD1 with greater affinity or potency, compared to LSD2. In general, selectivity can be at least about 5-fold, at least about 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold.
As inhibitors of LSD1, the compounds of the invention are useful in treating LSD1- 20  mediated diseases and disorders. The term "LSD1-mediated disease” or“LSD1-mediated
disorder" refers to any disease or condition in which LSD1 plays a role, or where the disease or condition is associated with expression or activity of LSD1. The compounds of the invention can    
therefore be used to treat or lessen the severity of diseases and conditions where LSD1 is known to play a role.
Diseases and conditions treatable using the compounds of the invention include generally cancers, inflammation, autoimmune diseases, viral induced pathogenesis, beta-globinopathies, 5  and other diseases linked to LSD1 activity.
Cancers treatable using compounds according to the present invention include, for example, hematological cancers, sarcomas, lung cancers, gastrointestinal cancers, genitourinary tract cancers, liver cancers, bone cancers, nervous system cancers, gynecological cancers, and skin cancers.
10  Example hematological cancers include, for example, lymphomas and leukemias such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma,
15  myeloproliferative diseases (e.g., primary myelofibrosis (PMF), polycythemia vera (PV),
essential thrombocytosis (ET)), myelodysplasia syndrome (MDS), and multiple myeloma.
Example sarcomas include, for example, chondrosarcoma, Ewing’s sarcoma, osteosarcoma, rhabdomyosarcoma, angiosarcoma, fibrosarcoma, liposarcoma, myxoma, rhabdomyoma, fibroma, lipoma, harmatoma, and teratoma.
20  Example lung cancers include, for example, non-small cell lung cancer (NSCLC),
bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, chondromatous hamartoma, and mesothelioma.
Example gastrointestinal cancers include, for example, cancers of the esophagus 25  (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), and 30  colorectal cancer.
   
Example genitourinary tract cancers include, for example, cancers of the kidney
(adenocarcinoma, Wilm's tumor [nephroblastoma]), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), and testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, 5  sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma).
Example liver cancers include, for example, hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, and hemangioma.
Example bone cancers include, for example, osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant 10  lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma, and giant cell tumors
Example nervous system cancers include, for example, cancers of the skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma,
15  meningiosarcoma, gliomatosis), brain (astrocytoma, meduoblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma,
retinoblastoma, congenital tumors), and spinal cord (neurofibroma, meningioma, glioma, sarcoma), as well as neuroblastoma and Lhermitte-Duclos disease.
Example gynecological cancers include, for example, cancers of the uterus (endometrial 20  carcinoma), cervix (cervical carcinoma, pre -tumor cervical dysplasia), ovaries (ovarian
carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma
25  (embryonal rhabdomyosarcoma), and fallopian tubes (carcinoma).
Example skin cancers include, for example, melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, and keloids.
The compounds of the invention can further be used to treat cancer types where LSD1 30  may be overexpressed including, for example, breast, prostate, head and neck, laryngeal, oral, and thyroid cancers (e.g., papillary thyroid carcinoma).    
The compounds of the invention can further be used to treat genetic disorders such as Cowden syndrome and Bannayan-Zonana syndrome.
The compounds of the invention can further be used to treat viral diseases such as herpes simplex virus (HSV), varicella zoster virus (VZV), human cytomegalovirus, hepatitis B virus 5  (HBV), and adenovirus.
The compounds of the invention can further be used to treat beta-globinopathies including, for example, beta-thalassemia and sickle cell anemia.
As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" a LSD1 protein with a 10  compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a LSD1 protein, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the LSD1 protein.
As used herein, the term "individual" or "patient, " used interchangeably, refers to any 15  animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine,
cattle, sheep, horses, or primates, and most preferably humans.
As used herein, the phrase "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, 20  medical doctor or other clinician.
As used herein, the term "treating" or "treatment" refers to inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e.,, arresting further development of the pathology and/or symptomatology) or ameliorating the disease; for 25  example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e.,, reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
As used herein, the term "preventing" or "prevention" refers to preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to 30  the disease, condition or disorder but does not yet experience or display the pathology or
symptomatology of the disease.    
Combination Therapies
The compounds of the invention can be used in combination treatments where the compound of the invention is administered in conjunction with other treatments such as the 5  administration of one or more additional therapeutic agents. The additional therapeutic agents are typically those which are normally used to treat the particular condition to be treated. The additional therapeutic agents can include, e.g., chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr-Abl, Flt-3, RAF, FAK, JAK, PIM, PI3K inhibitors for treatment of LSD1-mediated diseases, disorders or conditions. The one or more additional 10  pharmaceutical agents can be administered to a patient simultaneously or sequentially.
In some embodiments, the compounds of the invention can be used in combination with a therapeutic agent that targets an epigenetic regulator. Examples of epigenetic regulators include the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases. Histone deacetylase 15  inhibitors include, e.g., vorinostat.
For treating cancer and other proliferative diseases, the compounds of the invention can be used in combination with chemotherapeutic agents, agonists or antagonists of nuclear receptors, or other anti-proliferative agents. The compounds of the invention can also be used in combination with medical therapy such as surgery or radiotherapy, e.g., gamma-radiation, 20  neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and
systemic radioactive isotopes. Examples of suitable chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin,
25  carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide,
cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone
propionate, eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, 30  gefitinib, gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate,
ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan,    
lapatinib ditosylate, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone phenpropionate, nelarabine, nofetumomab, oxaliplatin, paclitaxel, pamidronate, panitumumab, pegaspargase, pegfilgrastim, 5  pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine,
rasburicase, rituximab, ruxolitinib, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, teniposide, testolactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, vorinostat, and zoledronate.
10  For treating cancer and other proliferative diseases, the compounds of the invention can be used in combination with ruxolitinib.
For treating autoimmune or inflammatory conditions, the compound of the invention can be administered in combination with a corticosteroid such as triamcinolone, dexamethasone, fluocinolone, cortisone, prednisolone, or flumetholone.
15  For treating autoimmune or inflammatory conditions, the compound of the invention can be administered in combination with an immune suppressant such as fluocinolone acetonide (Retisert®), rimexolone (AL-2178, Vexol, Alcon), or cyclosporine (Restasis®).
For treating autoimmune or inflammatory conditions, the compound of the invention can be administered in combination with one or more additional agents selected from Dehydrex™ 20  (Holles Labs), Civamide (Opko), sodium hyaluronate (Vismed, Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine, doxycycline (ALTY-0501, Alacrity), minocycline, iDestrin™ (NP50301, Nascent Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline25  (Duramycin, MOLI1901, Lantibio), CF101 (2S, 3S, 4R, 5R)-3, 4-dihydroxy-5-[6-[(3- iodophenyl)methylamino]purin-9-yl]-N-methyl-oxolane-2-carbamyl, Can-Fite Biopharma), voclosporin (LX212 or LX214, Lux Biosciences), ARG103 (Agentis), RX-10045 (synthetic resolvin analog, Resolvyx), DYN15 (Dyanmis Therapeutics), rivoglitazone (DE011, Daiichi Sanko), TB4 (RegeneRx), OPH-01 (Ophtalmis Monaco), PCS101 (Pericor Science), REV1-31 30  (Evolutec), Lacritin (Senju), rebamipide (Otsuka-Novartis), OT-551 (Othera), PAI-2 (University of Pennsylvania and Temple University), pilocarpine, tacrolimus, pimecrolimus (AMS981,    
Novartis), loteprednol etabonate, rituximab, diquafosol tetrasodium (INS365, Inspire), KLS- 0611 (Kissei Pharmaceuticals), dehydroepiandrosterone, anakinra, efalizumab,
mycophenolate sodium, etanercept (Embrel®), hydroxychloroquine, NGX267 (TorreyPines Therapeutics), or thalidomide.
5  For treating beta-thalassemia or sickle cell disease, the compound of the invention can be administered in combination with one or more additional agents such as Hydrea® (hydroxyurea).
In some embodiments, the compound of the invention can be administered in combination with one or more agents selected from an antibiotic, antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and non-steroidal anti-inflammatories, and anti- 10  allergic agents. Examples of suitable medicaments include aminoglycosides such as amikacin, gentamycin, tobramycin, streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin, norfloxacin, ofloxacin, trovafloxacin, lomefloxacin, levofloxacin, and enoxacin; naphthyridine; sulfonamides; polymyxin; chloramphenicol; neomycin; paramomycin;
colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its derivatives ("rifampins"); 15  cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole; flucytosine; natamycin; miconazole; ketoconazole; corticosteroids; diclofenac; flurbiprofen; ketorolac; suprofen;
cromolyn; lodoxamide; levocabastin; naphazoline; antazoline; pheniramine; or azalide antibiotic.
Other examples of agents, one or more of which a provided compound may also be combined with include: a treatment for Alzheimer's Disease such as donepezil and rivastigmine; 20  a treatment for Parkinson's Disease such as L-DOPA/carbidopa, entacapone, ropinirole,
pramipexole, bromocriptine, pergolide, trihexyphenidyl, and amantadine; an agent for treating multiple sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), glatiramer acetate, and mitoxantrone; a treatment for asthma such as albuterol and montelukast; an agent for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; an anti-inflammatory agent 25  such as a corticosteroid, such as dexamethasone or prednisone, a TNF blocker, IL-1 RA,
azathioprine, cyclophosphamide, and sulfasalazine; an immunomodulatory agent, including immunosuppressive agents, such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, an interferon, a corticosteroid, cyclophosphamide, azathioprine, and sulfasalazine; a neurotrophic factor such as an acetylcholinesterase inhibitor, an MAO inhibitor, an interferon, an anti- 30  convulsant, an ion channel blocker, riluzole, or an anti-Parkinson's agent; an agent for treating cardiovascular disease such as a beta-blocker, an ACE inhibitor, a diuretic, a nitrate, a calcium    
channel blocker, or a statin; an agent for treating liver disease such as a corticosteroid, cholestyramine, an interferon, and an anti-viral agent; an agent for treating blood disorders such as a corticosteroid, an anti-leukemic agent, or a growth factor; or an agent for treating immunodeficiency disorders such as gamma globulin.
Formulation, Dosage Forms and Administration
When employed as pharmaceuticals, the compounds of the invention can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending 10  upon whether local or systemic treatment is desired and upon the area to be treated.
Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous,
15  intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or
intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical 20  carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
This invention also includes pharmaceutical compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical administration. In making the compositions 25  of the invention, the active ingredient is typically mixed with an excipient, diluted by an
excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, 30  suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments
   
containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is
5  substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g., about 40 mesh.
The compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation 10  types. Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl 15  cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures 20  known in the art.
The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity 25  of active material calculated to produce the desired therapeutic effect, in association with a
suitable pharmaceutical excipient.
The active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, 30  according to the relevant circumstances, including the condition to be treated, the chosen route of
   
administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a 5  homogeneous mixture of a compound of the present invention. When referring to these
preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 10  about 0.1 to about 1000 mg of the active ingredient of the present invention.
The tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which 15  serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
The liquid forms in which the compounds and compositions of the present invention can 20  be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar
pharmaceutical vehicles.
Compositions for inhalation or insufflation include solutions and suspensions in
25  pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing 30  device can be attached to a face masks tent, or intermittent positive pressure breathing machine.
   
Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
Topical formulations can contain one or more conventional carriers. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, 5  for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white vaseline, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g., glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and 10  the like. In some embodiments, topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention. The topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
15  The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its
20  complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization 25  techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or
lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical 30  salts.
   
The therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition 5  can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 g/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from 10  about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
15  The compositions of the invention can further include one or more additional
pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed hereinabove. Labeled Compounds and Assay Methods
20  Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating LSD1 in tissue samples, including human, and for identifying LSD1 ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes LSD1 assays that contain such labeled compounds. 25  The present invention further includes isotopically-labeled compounds of the invention.
An "isotopically" or "radio-labeled" compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present
30  invention include but are not limited to 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 18F, 35S, 36Cl, 82Br, 75Br, 76Br, 77Br, 123I, 124I, 125I and 131I. The radionuclide that is    
incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound.
It is to be understood that a "radio-labeled " or "labeled compound" is a compound that has incorporated at least one radionuclide. In some embodiments the radionuclide is selected 5  from the group consisting of 3H, 14C, 125I, 35S and 82Br. In some embodiments, the compound incorporates 1, 2, or 3 deuterium atoms.
The present invention can further include synthetic methods for incorporating radio- isotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily 10  recognize the methods applicable for the compounds of invention.
A labeled compound of the invention can be used in a screening assay to
identify/evaluate compounds. For example, a newly synthesized or identified compound (i.e., test compound) which is labeled can be evaluated for its ability to bind LSD1 by monitoring its concentration variation when contacting with LSD1, through tracking of the labeling. For 15  example, a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to LSD1 (i.e., standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to LSD1directly correlates to its binding affinity. Conversely, in some other screening assays, the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the 20  labeled standard compound is monitored in order to evaluate the competition between the
standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the
25  invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters which can be changed or modified to yield essentially the same results. The compounds of the Examples were found to be inhibitors of LSD1 as described below. EXAMPLES
30  Experimental procedures for compounds of the invention are provided below.
Preparatory LC-MS purifications of some of the compounds prepared were performed on Waters    
mass directed fractionation systems. The basic equipment setup, protocols, and control software for the operation of these systems have been described in detail in the literature. See e.g.“Two- Pump At Column Dilution Configuration for Preparative LC-MS”, K. Blom, J. Combi. Chem., 4, 295 (2002);“Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis 5  Purification”, K. Blom, R. Sparks, J. Doughty, G. Everlof, T. Haque, A. Combs, J. Combi.
Chem., 5, 670 (2003); and "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", K. Blom, B. Glass, R. Sparks, A. Combs, J. Combi. Chem., 6, 874-883 (2004). The compounds separated were typically subjected to analytical liquid chromatography mass spectrometry (LCMS) for purity check under the following conditions: Instrument; Agilent 10  1100 series, LC/MSD, Column: Waters SunfireTM C18 5µm particle size, 2.1 x 5.0 mm, Buffers: mobile phase A: 0.025% TFA in water and mobile phase B: acetonitrile; gradient 2% to 80% of B in 3 minutes with flow rate 2.0 mL/minute.
Some of the compounds prepared were also separated on a preparative scale by reverse- phase high performance liquid chromatography (RP-HPLC) with MS detector or flash
15  chromatography (silica gel) as indicated in the Examples. Typical preparative reverse-phase high performance liquid chromatography (RP-HPLC) column conditions are as follows:
pH = 2 purifications: Waters SunfireTM C18 5 µm particle size, 19 x 100 mm column, eluting with mobile phase A: 0.1% TFA (trifluoroacetic acid) in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each 20  compound using the Compound Specific Method Optimization protocol as described in the
literature [see "Preparative LCMS Purification: Improved Compound Specific Method
Optimization", K. Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004)]. Typically, the flow rate used with the 30 x 100 mm column was 60 mL/minute.
pH = 10 purifications: Waters XBridge C18 5 µm particle size, 19 x 100 mm column, 25  eluting with mobile phase A: 0.15% NH4OH in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature [See
"Preparative LCMS Purification: Improved Compound Specific Method Optimization", K.
Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004)]. Typically, the flow 30  rate used with 30 x 100 mm column was 60 mL/minute.
   
Example 1
4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyridin-6- yl}benzonitrile
NC NH
5  Step 1: 4-(6-amino-2-chloropy
Figure imgf000053_0001
NC 2
A reaction vessel containin
Figure imgf000053_0002
6-chloropyridin-2-amine (415 mg, 2.00 mmol), (4-cyanophenyl)boronic acid (353 mg, 2.40 mmol), [1,1'- bis(diphenylphosphino)ferrocene]-dichloropalladium(II) complexed with dichloromethane (1:1) 10  (80 mg, 0.1 mmol) and potassium carbonate (550 mg, 4.0 mmol) in 1,4-dioxane (6 mL) and water (1 mL) was evacuated then refilled with nitrogen. The resulting mixture was heated to 80 oC and stirred for 3 h. The reaction mixture was cooled to room temperature then diluted with methylene chloride, washed with water, brine, dried over Na2SO4, filtered and concentrated. The residue was purified on a silica gel column eluting with 0 to 30 % EtOAc/DCM to give the 15  desired product as a white solid (320 mg, 71 %). LC-MS calculated for C12H9ClN3 (M+H)+: m/z = 230.0; found 230.0. Step 2: 4-[6-amino-2-(4-methylphenyl)pyridin-3-yl]benzonitrile
NC 2 20  A reaction vessel containin
Figure imgf000053_0003
o-2-chloropyridin-3-yl)benzonitrile (320 mg, 1.39 mmol), 4-methyl-8-(4-methylphenyl)-2,6-dioxotetrahydro[1,3,2]oxazaborolo[2,3- b][1,3,2]oxazaborol-4-ium-8-uide (413 mg, 1.67 mmol), [1,1'-    
bis(diphenylphosphino)ferrocene]dichloropalladium(II) complexed with dichloromethane (1:1) (60 mg, 0.07 mmol), and potassium carbonate (380 mg, 2.8 mmol) in 1,4-dioxane (5 mL) and water (1 mL) was evacuated then filled with nitrogen. The resulting mixture was heated to 110 oC and stirred overnight. The mixture was cooled to room temperature then diluted with 5  methylene chloride, washed with saturated NaHCO3 aqueous solution, water, brine, dried over Na2SO4, filtered and concentrated. The residue was purified on a silica gel column eluting with 0 to 30 % EtOAc/DCM to give the desired product as a light yellow solid (335 mg, 84 %). LC-MS calculated for C19H16N3 (M+H)+: m/z = 286.1; found 286.1. 10  Step 3: 4-[6-amino-5-bromo-2-(4-methylphenyl)pyridin-3-yl]benzonitrile
NC 2 To a mixture of 4-[6-amin
Figure imgf000054_0001
in-3-yl]benzonitrile (335 mg, 1.17 mmol) in tetrahydrofuran (5 mL) at 0 oC was added a solution of N-bromosuccinimide (230 mg, 1.3 mmol) in tetrahydrofuran (4 mL). The resulting yellow solution was stirred at 0 oC for 1.5 h 15  then diluted with methylene chloride, washed with saturated NaHCO3 aqueous solution, water, brine, dried over Na2SO4, filtered and concentrated. The residue was purified on a silica gel column eluting with 0 to 30 % EtOAc/DCM to give the desired product as a yellow solid (432 mg, quant.). LC-MS calculated for C19H15BrN3 (M+H)+: m/z = 364.0; found 364.0. 20  Step 4: N-[3-bromo-5-(4-cyanophenyl)-6-(4-methylphenyl)pyridin-2-yl]-N'- hydroxyimidoformamide
NC
H
To a mixture of 4-[6-amin
Figure imgf000054_0002
enyl)pyridin-3-yl]benzonitrile (275 mg, 0.755 mmol) in isopropyl alcohol (4 mL) was added 1,1-dimethoxy-N,N-    
dimethylmethanamine (0.20 mL, 1.5 mmol). The mixture was heated to 95 oC and stirred for 5 h. The resulting yellow solution was cooled to 50 oC then hydroxylamine hydrochloride (160 mg, 2.3 mmol) was added. The reaction mixture was stirred at 50 oC overnight then cooled to room temperature and concentrated. The residue was purified on a silica gel column eluting with 0 to 5  10 % MeOH/DCM to give the desired product as a yellow solid. LC-MS calculated for
C20H16BrN4O (M+H)+: m/z = 407.1; found 407.0. Step 5: 4-[8-bromo-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile
NC
r 10  To a solution of N-[3-bromo
Figure imgf000055_0001
(4-methylphenyl)pyridin-2-yl]-N'- hydroxyimidoformamide (307 mg, 0.754 mmol) in tetrahydrofuran (5 mL) at 0 oC was added trifluoroacetic anhydride (180 μL, 1.2 mmol). The resulting yellow solution was warmed to room temperature and stirred over t. The reaction was quenched with saturated NaHCO3 aqueous
Figure imgf000055_0002
solution then extracted with methylene chloride. The combined extracts were washed with water 15  and brine. The organic layer was dried over Na2SO4 then concentrated. The residue was purified on a silica gel column eluting with 0 to 20 % EtOAc/DCM to give the desired product as a yellow solid. LC-MS calculated for C20H14BrN4 (M+H)+: m/z = 389.0; found 389.1. Step 6: 4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyridin-6- 20  yl}benzonitrile
A mixture of 4-[8-bromo-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile (176 mg, 0.452 mmol), tert-butyl (3R)-3-(hydroxymethyl)pyrrolidine-1- carboxylate (182 mg, 0.904 mmol), π-allylpalladium chloride dimer (8 mg, 0.02 mmol), di-tert- butyl(2',4',6'-triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine (22 mg, 0.045 mmol) and 25  cesium carbonate (221 mg, 0.678 mmol) in toluene (6 mL) was evacuated then filled with
nitrogen. The resulting mixture was heated to 110 oC and stirred overnight. The reaction mixture was cooled to room temperature then diluted with water and extracted with EtOAc. The combined extracts were washed with water and brine. The organic layer was dried over Na2SO4    
then concentrated. The residue was purified on a silica gel column eluted with 0 to 50 %
EtOAc/DCM to give a yellow solid, which was dissolved in methylene chloride (1.5 mL) then trifluoroacetic acid (0.5 mL) was added. The resulting yellow solution was stirred at room temperature for 30 min then concentrated. The residue was dissolved in acetonitrile then purified 5  by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC- MS calculated for C25H24N5O (M+H)+: m/z = 410.2; found 410.2. Example 2
4-[5-(4-methylphenyl)-8-(2-pyrrolidin-3-ylethyl)[1,2,4]triazolo[1,5-a]pyridin-6- 10  yl]benzonitrile
NC NH
Step 1: tert-butyl 3-ethynylpyr
Figure imgf000056_0001
c To a solution of tert-butyl 3-for 1-carboxylate (580 mg, 2.91
Figure imgf000056_0002
15  mmol) in methanol (15 mL) at room temperature was added potassium carbonate (1.00 g, 7.28 mmol) and dimethyl (1-diazo-2-oxopropyl)phosphonate (839 mg, 4.37 mmol). The resulting mixture was stirred at room temperature for 3 h then passed through a short pad of celite and concentrated. The residue was purified on a silica gel column eluting with 0 to 50 %
EtOAc/Hexanes to give the product as a colorless oil which solidified upon standing in fridge to 20  give a white solid (374 mg, 66 %). Step 2: tert-butyl 3-{[6-(4-cyanophenyl)-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-8- yl]ethynyl}pyrrolidine-1-carboxylate
   
NC NBoc
A mixture of 4-[8-bro
Figure imgf000057_0001
lo[1,5-a]pyridin-6- yl]benzonitrile (Example 1, Step 5, 70. mg, 0.18 mmol), tert-butyl 3-ethynylpyrrolidine-1- carboxylate (53 mg, 0.27 mmol), tetrakis(triphenylphosphine)palladium(0) (21 mg, 0.018 5  mmol), and copper(I) iodide (6.8 mg, 0.036 mmol) in N,N-dimethylformamide (2 mL) was evacuated then filled with nitrogen. Then N,N-diisopropylethylamine (94 μL, 0.54 mmol) was added. The resulting mixture was heated to 85 oC and stirred for 4 h. T eaction mixture was
Figure imgf000057_0002
cooled to room temperature then diluted with EtOAc and washed with water and brine. The organic layer was dried over Na2SO4 then concentrated. The residue was purified on a silica gel 10  column eluting with 0 to 50 % EtOAc/DCM to give the desired product (62 mg, 68 %). LC-MS calculated for C31H30N5O2 (M+H)+: m/z = 504.2; found 504.2. Step 3: 4-[5-(4-methylphenyl)-8-(2-pyrrolidin-3-ylethyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile
15  To a solution of tert-butyl 3-{[6-(4-cyanophenyl)-5-(4-methylphenyl)[1,2,4]triazolo[1,5- a]pyridin-8-yl]ethynyl}pyrrolidine-1-carboxylate (62 mg, 0.12 mmol) in tetrahydrofuran (3 mL) and methanol (3 mL) was added palladium (10 wt% on activated carbon, 26 mg, 0.025 mmol). The resulting mixture was stirred under a balloon of hydrogen overnight. The mixture was filtered through a short pad of celite then washed with THF. The filtrate was concentrated 20  and the residue was dissolved in 3 mL of DCM then 1 mL of TFA was added. The resulting yellow solution was stirred at room temperature for 1 h then concentrated. The residue was dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C26H26N5 (M+H)+: m/z = 408.2; found 408.2.
25 
Example 3
4-{5-(4-methylphenyl)-8-[2-(1-methylpyrrolidin-3-yl)ethyl][1,2,4]triazolo[1,5-a]pyridin-6- yl}benzonitrile
   
NC
N
To a solution of 4-[5-(
Figure imgf000058_0001
-ylethyl)[1,2,4]triazolo[1,5- a]pyridin-6-yl]benzonitrile (Example 2, Step 3, 14 mg, 0.034 mmol) in tetrahydrofuran (2 mL) was added formaldehyde (37 wt% in water, 13 μL, 0.17 mmol), followed by acetic acid (5.8 5  μL, 0.10 mmol). The resulting solution was stirred at room temperature for 2 h, then sodium triacetoxyborohydride (22 mg, 0.10 mmol) was added. The reaction mixture was stirred at room temperature overnight then filtered and purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C27H28N5 (M+H)+: m/z = 422.2; found 422.3.
10 
Example 4
4-[8-{3-[(methylamino)methyl]phenyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile
NC
H
15  Step 1: 4-[8-(3-formylphenyl)-
Figure imgf000058_0002
[1,5-a]pyridin-6-yl]benzonitrile
NC
A mixture of 4-[8-bro
Figure imgf000058_0003
olo[1,5-a]pyridin-6- yl]benzonitrile (Example 1, Step 5, 53 mg, 0.14 mmol), (3-formylphenyl)boronic acid (41 mg, 0.27 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) complexed with20  dichloromethane (1:1) (10 mg, 0.01 mmol), and potassium carbonate (38 mg, 0.27 mmol) in 1,4- dioxane (2 mL) and water (0.2 mL) was evacuated then filled with nitrogen. The resulting mixture was heated to 90 oC and stirred for 6 h. The reaction mixture was cooled to room    
temperature then diluted with DCM, filtered and concentrated. The residue was purified on a silica gel column eluting with 0 to 20 % EtOAc/DCM to give the desired product as a yellow solid (45 mg, 80 %). LC-MS calculated for C27H19N4O (M+H)+: m/z = 415.2; found 415.2. 5  Step 2: 4-[8-{3-[(methylamino)methyl]phenyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin- 6-yl]benzonitrile
To a solution of 4-[8-(3-formylphenyl)-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin- 6-yl]benzonitrile (15 mg, 0.036 mmol) in tetrahydrofuran (2 mL) was added methylamine (2M in THF, 90. μL, 0.18 mmol), followed by acetic acid (10. μL, 0.18 mmol). The resulting mixture 10  was stirred at room temperature for 2 h, then sodium triacetoxyborohydride (23 mg, 0.11
mmol) was added. The reaction mixture was stirred at room temperature overnight then diluted with THF, filtered and purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C28H24N5 (M+H)+: m/z = 430.2; found 430.2.
15 
Example 5
4-[8-{3-[(3-aminopyrrolidin-1-yl)methyl]phenyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5- a]pyridin-6-yl]benzonitrile
NC NH 2 20  To a solution of 4-
Figure imgf000059_0001
,2,4]triazolo[1,5-a]pyridin- 6-yl]benzonitrile (Example 4, Step 1, 15 mg, 0.036 mmol) in tetrahydrofuran (2 mL) was added tert-butyl pyrrolidin-3-ylcarbamate (20 mg, 0.11 mmol), followed by acetic acid (10. μL, 0.18 mmol). The resulting mixture was stirred at room temperature for 2 h, then sodium
triacetoxyborohydride (23 mg, 0.11 mmol) was added. The reaction mixture was stirred at room 25  temperature overnight. The mixture was diluted with DCM then washed with saturated NaHCO3 aqueous solution. The organic layer was dried over Na2SO4 then concentrated. The residue was dissolved in methylene chloride (1 mL) then trifluoroacetic acid (1 mL) was added. The resulting yellow solution was stirred at room temperature for 1 h then concentrated. The residue was    
dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C31H29N6 (M+H)+: m/z = 485.2; found 485.3. 5  Example 6
4-[5-(4-methylphenyl)-8-(piperazin-1-ylmethyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile
NC H Step 1: 4-[5-(4-methylphenyl)-8
Figure imgf000060_0001
idin-6-yl]benzonitrile
NC
10 
A reaction vessel containin
Figure imgf000060_0002
o-5-(4- methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile (Example 1, Step 5, 338 mg, 0.868 mmol), 4-methyl-2,6-dioxo-8-vinyltetrahydro[1,3,2]oxazaborolo[2,3-b][1,3,2]oxazaborol-4-ium- 8-uide (206 mg, 1.13 mmol), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) 15  complexed with dichloromethane (1:1 ) (42 mg, 0.052 mmol), and potassium carbonate (240 mg, 1.7 mmol) in 1,4-dioxane (6 mL) and water (2 mL) was evacuated then filled with nitrogen. The resulting mixture was heated to 95 oC and stirred for 2 h. The mixture was cooled to room temperature then diluted with methylene chloride, washed with saturated NaHCO3 aqueous solution, dried over Na2SO4, filtered and concentrated. The residue was purified on a silica 20  gel column eluting with 0 to 30 % EtOAc/DCM to give the desired product as a yellow solid (225 mg, 77 %). LC-MS calculated for C22H17N4 (M+H)+: m/z = 337.1; found 337.1. Step 2: 4-[8-formyl-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile
   
NC
O H
To a solution of 4-[5-(4-me
Figure imgf000061_0001
4]triazolo[1,5-a]pyridin-6- yl]benzonitrile (225 mg, 0.669 mmol) in 1,4-dioxane (10 mL) and water (3 mL) was added osmium tetraoxide (4 wt% in water, 420 μL, 0.067 mmol). The resulting mixture was stirred at 5  room temperature for 10 min then sodium periodate (429 mg, 2.01 mmol) was added. The
reaction mixture was stirred at room temperature overnight. The reaction mixture was quenched with water then extracted with DCM. The combined extracts were washed with water and brine then dried over Na2SO4 and concentrated. The residue was purified on a silica gel column eluting with 0 to 30 % EtOAc/DCM to give the desired product as a yellow solid (159 mg, 70 %). LC- 10  MS calculated for C21H15N4O (M+H)+: m/z = 339.1; found 339.2. Step 3: 4-[5-(4-methylphenyl)-8-(piperazin-1-ylmethyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile
To a solution of 4-[8-formyl-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin-6- 15  yl]benzonitrile (14 mg, 0.041 mmol) in methylene chloride (2 mL) was added tert-butyl
piperazine-1-carboxylate (23 mg, 0.12 mmol), followed by acetic acid (12 μL, 0.21 mmol). The resulting mixture was stirred at room temperature overnight then sodium triacetoxyborohydride (26 mg, 0.12 mmol) was added. The reaction mixture was stirred at room temperature for 2 h then diluted with DCM and washed with saturated NaHCO3 aqueous solution. The organic layer 20  was dried over Na2SO4 then concentrated. The residue was dissolved in methylene chloride (1 mL) then trifluoroacetic acid (1 mL) was added. The resulting yellow solution was stirred at room temperature for 1 h then concentrated. The residue was dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C25H25N6 (M+H)+: m/z = 409.2; found 409.2.
25 
Example 7
4-{5-(4-methylphenyl)-8-[(4-methylpiperazin-1-yl)methyl][1,2,4]triazolo[1,5-a]pyridin-6- yl}benzonitrile     NC
To a solution of 4-[8-fo
Figure imgf000062_0001
triazolo[1,5-a]pyridin-6- yl]benzonitrile (Example 6, Step 2, 14 mg, 0.041 mmol) in methylene chloride (2 mL) was added 1-methyl-piperazine (14 μL, 0. 12 mmol), followed by acetic acid (12 μL, 0.21 mmol). The 5  resulting mixture was stirred at room temperature overnight then sodium triacetoxyborohydride (26 mg, 0.12 mmol) was added. The reaction mixture was stirred at room temperature for 2 h then diluted with DCM and washed with saturated NaHCO3 aqueous solution. The organic layer was dried over Na2SO4 then concentrated. The residue was dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC- 10  MS calculated for C26H27N6 (M+H)+: m/z = 423.2; found 423.3. Example 8
4-[8-{[(3S)-3-(dimethylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)[1,2,4]triazolo- [1,5-a]pyridin-6-yl]benzonitrile
NC
15 
To a solution of 4-[8-fo
Figure imgf000062_0002
triazolo[1,5-a]pyridin-6- yl]benzonitrile (Example 6, Step 2, 57 mg, 0.17 mmol) in methylene chloride (3.0 mL) was added (3S)-N,N-dimethylpyrrolidin-3-amine (TCI, Cat#D2193: 64 μL, 0.50 mmol), followed by acetic acid (28 μL, 0.50 mmol). The resulting mixture was stirred at room temperature for 1 h, 20  then sodium etoxyborohydride (71 mg, 0.34 mmol) was added. The reaction mixture was stirred at room temperature for 2 h then diluted with DCM and washed with saturated NaHCO3 aqueous solution. The organic layer was dried over Na2SO4 then concentrated. The residue was dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LCMS calculated for C27H29N6 (M+H)+: m/z = 437.2;
25  Found: 437.2. 1H NMR (500 MHz, DMSO) δ 8.53 (s, 1H), 7.86 (s, 1H), 7.79 (d, J = 8.3 Hz, 2H),    
7.41 (d, J = 8.3 Hz, 2H), 7.28 (d, J = 8.1 Hz, 2H), 7.22 (d, J = 8.1 Hz, 2H), 4.49 (s, 2H), 3.97 (br, 1H), 3.38 (br, 2H), 3.27 (br, 1H), 3.09 (br, 1H), 2.78 (s, 6H), 2.33 (s, 3H), 2.29 (br, 1H), 2.19– 2.08 (m, 1H). 5  Example 9
4-[8-{[(3R)-3-(dimethylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)[1,2,4]triazolo- [1,5-a]pyridin-6-yl]benzonitrile
NC
This compound was pre
Figure imgf000063_0001
us to those described for Example 10  8 with (3R)-N,N-dimethylpyrrolidin-3-amine replacing (3S)-N,N-dimethylpyrrolidin-3-amine.
The compound was purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LCMS calculated for C27H29N6 (M+H)+: m/z = 437.2; Found: 436.7. Example 10
15  4-[8-{[(3S)-3-(methylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5- a]pyridin-6-yl]benzonitrile
NC
To a solution of 4-[8-fo
Figure imgf000063_0002
triazolo[1,5-a]pyridin-6- yl]benzonitrile (Example 6, Step 2, 6.0 mg, 0.018 mmol) in methylene chloride (1.0 mL) was 20  added tert-butyl methyl[(3S)-pyrrolidin-3-yl]carbamate (18 mg, 0.089 mmol), followed by acetic acid (10 μL, 0.18 mmol). The resulting mixture was stirred at room temperature overnight, then sodiu cetoxyborohydride (11 mg, 0.053 mmol) was added. The reaction mixture was stirred at room temperature for 2 h then diluted with DCM and washed with saturated NaHCO3 aqueous solution. The organic layer was dried over Na2SO4 then concentrated. The residue was dissolved 25  in methylene chloride (1 mL) then trifluoroacetic acid (0.5 mL) was added. The resulting yellow    
solution was stirred at room temperature for 2 h then concentrated. The residue was dissolved in acetonitrile then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LCMS calculated for C26H27N6 (M+H)+: m/z = 423.2; Found: 423.2. 5  Example 11
4-[8-{[(3R)-3-(methylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5- a]pyridin-6-yl]benzonitrile
NC
This compound was pre
Figure imgf000064_0001
us to those described for Example10  10 with tert-butyl methyl[(3R)-pyrrolidin-3-yl]carbamate replacing tert-butyl methyl[(3S)- pyrrolidin-3-yl]carbamate. The compound was purified by prep HPLC (pH = 2,
acetonitrile/water+TFA) to give the desired product as the TFA salt. LCMS calculated for C26H27N6 (M+H)+: m/z = 423.2; Found: 423.1. 15  Example 12
4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyrazin-6- yl}benzonitrile
NC NH
Step 1: 4-(5-amino-3-chlorop
Figure imgf000064_0002
NC
20  2
A mixture of 5-bromo-6-ch 04 g, 5.00 mmol), (4-
Figure imgf000064_0003
cyanophenyl)boronic acid (0.882 g, 6.00 mmol), dichloro(bis{di-tert-butyl[4-    
(dimethylamino)phenyl]phosphoranyl})palladium (110 mg, 0.15 mmol), sodium carbonate (1.06 g, 10.0 mmol) in 1,4-dioxane (12.0 mL) and water (2.0 mL) was evacuated then filled with nitrogen. The resulting mixture was stirred at 90 oC for 4 h then cooled to room temperature. The mixture was diluted with methylene chloride (15 mL) and water (5 mL). The precipitates 5  were collected by filtration and washed with methyl t-butyl ether then dried to afford the desired product (1.05 g, 91 %). LC-MS calculated for C11H8ClN4 (M+H)+: m/z = 231.0; found 231.1. Step 2: 4-[5-amino-3-(4-methylphenyl)pyrazin-2-yl]benzonitrile
NC 2 10  A reaction vessel containin
Figure imgf000065_0001
o-3-chloropyrazin-2-yl)benzonitrile (1.15 g, 5.00 mmol), (4-methylphenyl)boronic acid (0.86 g, 6.4 mmol), sodium carbonate (1.06 g, 10.0 mmol), and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) complexed with dichloromethane (1:1) (0.20 g, 0.25 mmol) in 1,4-dioxane (20.0 mL) and water (4.0 mL) was evacuated then refilled with nitrogen. The resulting mixture was stirred at 110 °C for 3 h then 15  cooled to room temperature. The mixture was diluted with methylene chloride, washed with saturated NaHCO3 aqueous solution, water and brine. The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was treated with DCM/diethyl- ether (1:1). The precipitate was collected by filtration to afford the desired product (0.61g). The filtrate was concentrated and the residue was purified by flash chromatography on a silica gel 20  column eluting with 0 to 100 % EtOAc/DCM to afford another batch of the product (0.60 g).
LC-MS calculated for C18H15N4 (M+H)+: m/z = 287.1; found 287.1. Step 3: 4-[5-amino-6-bromo-3-(4-methylphenyl)pyrazin-2-yl]benzonitrile
NC 2
Figure imgf000065_0002
   
To a solution of 4-[5-amino-3-(4-methylphenyl)pyrazin-2-yl]benzonitrile (2.40 g, 8.38 mmol) in tetrahydrofuran (36 mL) at 0 oC was added N-bromosuccinimide (1.64 g, 9.22 mmol). The resulting mixture was stirred at 0 oC for 1 h then warmed to room temperature. The mixture was diluted with methylene chloride, washed with saturated NaHCO3 aqueous solution, water, 5  and brine. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified on a silica gel column eluting with 0 to 60 % EtOAc/DCM to give the desired product (2.8 g, 92 %). LC-MS calculated for C18H14BrN4 (M+H)+: m/z = 365.0; found 365.0. Step 4: tert-butyl (3R)-3-({[3-amino-6-(4-cyanophenyl)-5-(4-methylphenyl)pyrazin-2- 10  yl]oxy}methyl)pyrrolidine-1-carboxylate
NC NBoc
To a solution of tert-
Figure imgf000066_0001
idine-1-carboxylate (2.06 g, 10.2 mmol) in tetrahydrofuran (25 mL) at room temperature was added NaH (60 wt.% in mineral oil, 413 mg, 17.2 mmol). The resulting mixture was stirred at room temperature for 30 min then 4- 15  [5-amino-6-bromo-3-(4-methylphenyl)pyrazin-2-yl]benzonitrile (1.50 g, 4.10 mmol) was added.
The reaction mixture was stirred at 85 °C for 15 h then cooled to room temperature. The mixture was quenched with saturated NaHCO3 aqueous solution and extracted with EtOAc. The combined organic layers were dried over Na2SO4, and concentrated. The residue was purified on a silica gel column eluting with 10 to 40 % EtOAc/DCM to give the product as a yellow solid. 20  LC-MS calculated for C24H24N5O3 (M-tBu+2H)+: m/z = 430.2; found 430.1. Step 5: 4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyrazin-6- yl}benzonitrile
A mixture of tert-butyl (3R)-3-({[3-amino-6-(4-cyanophenyl)-5-(4-25  methylphenyl)pyrazin-2-yl]oxy}methyl)pyrrolidine-1-carboxylate (100 mg, 0.2 mmol) and 1,1- dimethoxy-N,N-dimethylmethanamine (137 μL, 1.03 mmol) in isopropyl alcohol (1.5 mL) was heated to 95 °C and stirred for 2 h. The reaction mixture was cooled to room temperature then concentrated. The residue was dissolved in methanol (1.5 mL) and cooled to 0 °C then pyridine    
(50. μL, 0.62 mmol) was added, followed by hydroxylamine-O-sulfonic acid (58 mg, 0.51 mmol). The reaction mixture was warmed to room temperature and stirred overnight. The mixture was then quenched with saturated NaHCO3 solution and extracted with EtOAc. The combined extracts were dried over Na2SO4 and then concentrated. The residue was purified on a 5  silica gel column to give the desired intermediate, which was then dissolved in methylene
chloride (1.5 mL) and trifluoroacetic acid (0.5 mL) was added. The mixture was stirred at room temperature for 1 h and then concentrated. The crude material was then purified by prep-HPLC (pH = 2, acetonitrile/water+TFA) to give the desired product as the TFA salt. LC-MS calculated for C24H23N6O (M+H)+: m/z = 411.2; found 411.2. 1H NMR (500 MHz, DMSO) δ 8.86 (br, 2H), 10  8.61 (s, 1H), 7.80– 7.72 (m, 2H), 7.57– 7.51 (m, 2H), 7.31 (d, J = 8.1 Hz, 2H), 7.26 (d, J = 8.1 Hz, 2H), 4.69– 4.56 (m, 2H), 3.48– 3.38 (m, 1H), 3.38– 3.18 (m, 2H), 3.16– 3.06 (m, 1H), 2.98– 2.87 (m, 1H), 2.35 (s, 3H), 2.22– 2.12 (m, 1H), 1.91– 1.80 (m, 1H). Example 13
15  4-(5-(4-methylphenyl)-8-{[(3R)-1-methylpyrrolidin-3-yl]methoxy}[1,2,4]triazolo[1,5- a]pyrazin-6-yl)benzonitrile
NC
N
 
To a solution of 4-{5-(
Figure imgf000067_0001
in-3- ylmethoxy][1,2,4]triazolo[1,5-a]pyrazin-6-yl}benzonitrile (Example 12: 10 mg, 0.02 mmol) 20  in methylene chloride (1.5 mL) was added formaldehyde (37 wt.% in water, 18.1 μL, 0.244 mmol), followed by acetic acid (6.9 μL, 0.12 mmol). The resulting mixture was stirred at room temperature for 3 h then sodium triacetoxyborohydride (26 mg, 0.12 mmol) was added.
The reaction mixture was stirred at room temperature for another 2 h then concentrated. The resulting residue was then purified by prep HPLC (pH = 2, acetonitrile/water+TFA) to give the 25  desired product as the TFA salt. LC-MS calculated for C25H25N6O (M+H)+: m/z = 425.2; found 425.2.
    Example A: LSD1 histone demethylase biochemical assay
LANCE LSD1/KDM1A demethylase assay- 10 μL of 1 nM LSD-1 enzyme (ENZO BML-SE544-0050) in the assay buffer (50 mM Tris, pH 7.5, 0.01% Tween-20, 25 mM NaCl, 5 mM DTT) were preincubated for 1 hour at 25oC with 0.8 uL compound/DMSO dotted in black 5  384 well polystyrene plates. Reactions were started by addition of 10 μL of assay buffer
containing 0.4 μM Biotin-labeled Histone H3 peptide substrate: ART-K(Me1)- QTARKSTGGKAPRKQLA-GGK(Biotin) SEQ ID NO:1 (AnaSpec 64355) and incubated for 1 hour at 25oC. Reactions were stopped by addition of 10 μL 1 X LANCE Detection Buffer (PerkinElmer CR97-100) supplemented with 1.5 nM Eu-anti-unmodified H3K4 Antibody 10  (PerkinElmer TRF0404), and 225 nM LANCE Ultra Streptavidin (PerkinElmer TRF102) along with 0.9 mM Tranylcypromine-HCl (Millipore 616431). After stopping the reactions plates were incubated for 30 minutes and read on a PHERAstar FS plate reader (BMG Labtech). IC50 data for the example compounds is provided in Table 1 (+ refers to IC50≤ 50 nM; ++ refers to IC50 > 50 nM and≤ 100 nM; +++ refers to IC50 > 50 nM and≤ 100 nM; ++++ refers to IC50 > 500 nM 15  and≤ 1000 nM).
Table 1
Figure imgf000068_0001
Various mod ed herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also 20  intended to fall within the scope of the appended claims. Each reference, including all patent,
   
patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.
   

Claims

What is claimed is:
1. A compound of Formula I:
Figure imgf000070_0001
or a pharmaceutically acceptable salt thereof, wherein:
X is N or CRX;
Ring A is C6-10 aryl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA;
Ring B is C6-10 aryl; 5-10 membered heteroaryl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; C3-10 cycloalkyl; or 4-10 membered heterocycloalkyl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; wherein said C6-10 aryl, 5-10 membered heteroaryl, C3-10 cycloalkyl, and 4-10 membered heterocycloalkyl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, or
S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and
S(O)2NRc1Rd1;
wherein when X is CRX, then R1 is not CN;
   
R2 is H, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy2, CN, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, or
S(O)2NRc2Rd2; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy2, halo, CN, ORa2, SRa2, C(O)Rb2, C(O)NRc2Rd2, C(O)ORa2, OC(O)Rb2, OC(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, NRc2C(O)ORa2, NRc2C(O)NRc2Rd2, C(=NRe2)Rb2, C(=NRe2)NRc2Rd2, NRc2C(=NRe2)NRc2Rd2, NRc2S(O)Rb2, NRc2S(O)2Rb2, NRc2S(O)2NRc2Rd2, S(O)Rb2, S(O)NRc2Rd2, S(O)2Rb2, and
S(O)2NRc2Rd2;
each RA is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, OC(O)NRc4Rd4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4C(O)NRc4Rd4, C(=NRe4)Rb4, C(=NRe4)NRc4Rd4, NRc4C(=NRe4)NRc4Rd4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4,
S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3, substituents independently selected from halo, C1-6 haloalkyl, CN, NO2, ORa4, SRa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, OC(O)Rb4, OC(O)NRc4Rd4, NRc4Rd4, NRc4C(O)Rb4, NRc4C(O)ORa4, NRc4C(O)NRc4Rd4, C(=NRe4)Rb4, C(=NRe4)NRc4Rd4, NRc4C(=NRe4)NRc4Rd4, NRc4S(O)Rb4, NRc4S(O)2Rb4, NRc4S(O)2NRc4Rd4, S(O)Rb4, S(O)NRc4Rd4, S(O)2Rb4, and S(O)2NRc4Rd4;
each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5,
OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(=NRe5)Rb5, C(=NRe5)NRc5Rd5, NRc5C(=NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3 substituents independently selected from Cy3, halo, C1-6 haloalkyl, CN, NO2, ORa5, SRa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, OC(O)Rb5, OC(O)NRc5Rd5, NRc5Rd5, NRc5C(O)Rb5, NRc5C(O)ORa5, NRc5C(O)NRc5Rd5, C(=NRe5)Rb5, C(=NRe5)NRc5Rd5, NRc5C(=NRe5)NRc5Rd5, NRc5S(O)Rb5, NRc5S(O)2Rb5, NRc5S(O)2NRc5Rd5, S(O)Rb5, S(O)NRc5Rd5, S(O)2Rb5, and S(O)2NRc5Rd5;
   
RX is independently selected from H, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)ORa7, NRc7C(O)NRc7Rd7, C(=NRe7)Rb7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, NRc7S(O)Rb7, NRc7S(O) 7
2Rb7, NRc7S(O)2NRc7Rd , S(O)Rb7,
S(O)NRc7Rd7, S(O)2Rb7, and S(O)2NRc7Rd7;
each Cy1, Cy2, Cy3, and Cy4 is independently selected from C6-10 aryl, C3-10 cycloalkyl, 5- 10 membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6,
NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6,
NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and
S(O)2NRc6Rd6, wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted by 1, 2, or 3 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, and Cy4; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, SRa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, OC(O)Rb3, OC(O)NRc3Rd3, NRc3Rd3, NRc3C(O)Rb3, NRc3C(O)ORa3,
NRc3C(O)NRc3Rd3, C(=NRe3)Rb3, C(=NRe3)NRc3Rd3, NRc3C(=NRe3)NRc3Rd3, NRc3S(O)Rb3, NRc3S(O)2Rb3, NRc3S(O)2NRc3Rd3, S(O)Rb3, S(O)NRc3Rd3, S(O)2Rb3, and S(O)2NRc3Rd3;
each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered    
heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra2, Rb2, Rc2, and Rd2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,    
NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc2 and Rd2 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc3 and Rd3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,    
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra4, Rb4, Rc4, and Rd4 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc4 and Rd4 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra5, Rb5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,
   
NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc5 and Rd5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc6 and Rd6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra7, Rb7, Rc7, and Rd7 is independently selected from H, C1-4 alkyl, C1-4 haloalkyl, C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino,    
halo, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, C1-4 alkylamino, di(C1-4 alkyl)amino, C1-4 haloalkyl, and C1-4 haloalkoxy; and
each Re1, Re2, Re3, Re4, Re5, Re6, and Re7 is independently selected from H, C1-4 alkyl, and CN. 2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CRX;
Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA;
Ring B is phenyl or 5-6 membered heteroaryl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; wherein said phenyl and 5-6 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, or
S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, SRa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, OC(O)Rb1, OC(O)NRc1Rd1, NRc1Rd1, NRc1C(O)Rb1,
NRc1C(O)ORa1, NRc1C(O)NRc1Rd1, C(=NRe1)Rb1, C(=NRe1)NRc1Rd1, NRc1C(=NRe1)NRc1Rd1, NRc1S(O)Rb1, NRc1S(O)2Rb1, NRc1S(O)2NRc1Rd1, S(O)Rb1, S(O)NRc1Rd1, S(O)2Rb1, and
S(O)2NRc1Rd1;
wherein when X is CRX, then R1 is not CN;
R2 is H, halo, C1-6 alkyl, CN, ORa2, C(O)Rb2, C(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, S(O)2Rb2, or S(O)2NRc2Rd2; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from halo, CN, ORa2, C(O)Rb2, C(O)NRc2Rd2, NRc2Rd2, NRc2C(O)Rb2, S(O)2Rb2, and S(O)2NRc2Rd2;
each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected    
from halo, C 4 1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa , NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4;
each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5, NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1 , 2, or 3 substituents independently selected from Cy3, halo, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5, NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5;
RX is independently selected from H, halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, NRc7Rd7, NRc7C(O)Rb7, S(O)2Rb7, and S(O)2NRc7Rd7;
each Cy1, Cy3, and Cy4 is independently selected from C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, and 4-10 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6,
NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6,
NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and
S(O)2NRc6Rd6, wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted by 1, 2, or 3 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, C(=NRe6)NRc6Rd6, NRc6C(=NRe6)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, and Cy4; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3;    
each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra2, Rb2, Rc2, and Rd2 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with    
1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc2 and Rd2 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, phenyl, 5-6 membered heteroaryl, C 7
1-6 haloalkyl, halo, CN, ORa , SRa7, C(O)Rb7, C(O)NRc7Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1,
2,
3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc3 and Rd3 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl,
4-7 membered heterocycloalkyl, phenyl,    
5-6 membered heteroaryl, C 7
1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc Rd7,
C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, phenyl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra4, Rb4, Rc4, and Rd4 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc4 and Rd4 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra5, Rb5, Rc5, and Rd5 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C6-10 aryl, C3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C6-10 aryl-C1-4 alkyl-, C3-10 cycloalkyl-C1-4 alkyl-, (5-10 membered heteroaryl)- C1-4 alkyl-, and (4-10 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl,    
halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc5 and Rd5 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4- 7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7,
NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1- 4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7,
OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7,
C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc6 and Rd6 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7,
C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7,
NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, C(=NRe7)NRc7Rd7, NRc7C(=NRe7)NRc7Rd7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra7, Rb7, Rc7, and Rd7 is independently selected from H, C1-4 alkyl, C1-4 haloalkyl, C2-4 alkenyl, and C2-4 alkynyl, wherein said C1-4 alkyl, C2-4 alkenyl, and C2-4 alkynyl are each    
optionally substituted with 1, 2, or 3 substituents independently selected from OH, CN, amino, halo, C1-4 alkyl, C1-4 alkoxy, C1-4 alkylthio, C1-4 alkylamino, di(C1-4 alkyl)amino, C1-4 haloalkyl, and C1-4 haloalkoxy; and
each Re1, Re6, and Re7 is independently selected from H, C1-4 alkyl, and CN. 3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
X is N or CRX;
Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from RA;
Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected from RB;
R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, S(O)2Rb1, or S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, S(O)2Rb1, and S(O)2NRc1Rd1;
R2 is H;
each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, and ORa4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from CN and ORa4;
each RB is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, and ORa5;
RX is H;
each Cy1 and Cy4 is independently selected from phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy;
each RCy is independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, C2-6 alkenyl, C2-6 alkynyl, CN, NO2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NRc6Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6, NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6, wherein said C1-4 alkyl, C2-6 alkenyl, C2-6 alkynyl, are each optionally substituted by 1, 2, or 3    
substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, NO c6
2, ORa6, SRa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, OC(O)Rb6, OC(O)NR Rd6, NRc6Rd6, NRc6C(O)Rb6, NRc6C(O)ORa6, NRc6C(O)NRc6Rd6, NRc6S(O)Rb6, NRc6S(O)2Rb6,
NRc6S(O)2NRc6Rd6, S(O)Rb6, S(O)NRc6Rd6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Ra1 is independently selected from H, C1-6 alkyl, and 4-7 membered
heterocycloalkyl; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3, and wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, S(O)2Rb6, and S(O)2NRc6Rd6;
each Rb1, Rc1, and Rd1 is independently selected from H, C1-6 alkyl, C1-4 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl-, wherein said C1-6 alkyl, C2-6 alkenyl, C2- 6 alkynyl, phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, 4-7 membered heterocycloalkyl, phenyl-C1-4 alkyl-, C3-7 cycloalkyl-C1-4 alkyl-, (5-6 membered heteroaryl)-C1-4 alkyl-, and (4-7 membered heterocycloalkyl)-C1-4 alkyl- are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7,
NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7,
NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
or any Rc1 and Rd1 together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 substituents independently selected from C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, 5-6 membered heteroaryl, C1-6 haloalkyl, halo, CN, ORa7, SRa7, C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7,
NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7, NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7, wherein said C1-6 alkyl, C3-7 cycloalkyl, 4-7 membered heterocycloalkyl, C6-10 aryl, and 5-6 membered heteroaryl are each optionally substituted by 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, ORa7, SRa7,    
C(O)Rb7, C(O)NRc7Rd7, C(O)ORa7, OC(O)Rb7, OC(O)NRc7Rd7, NRc7Rd7, NRc7C(O)Rb7, NRc7C(O)NRc7Rd7, NRc7C(O)ORa7, S(O)Rb7, S(O)NRc7Rd7, S(O)2Rb7, NRc7S(O)2Rb7,
NRc7S(O)2NRc7Rd7, and S(O)2NRc7Rd7;
each Ra3, Rb3, Rc3, and Rd3 is independently selected from H and C1-6 alkyl;
each Ra4 is independently selected from H and C1-6 alkyl;
each Ra5 is independently selected from H and C1-6 alkyl;
each Ra6, Rb6, Rc6, and Rd6 is independently selected from H and C1-6 alkyl; and each Ra7, Rb7, Rc7, and Rd7 is independently selected from H and C1-4 alkyl. 4. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein X is N. 5. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein X is CRX.
6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl or 5-10 membered heteroaryl comprising carbon and 1, 2, 3, or 4 heteroatoms selected from N, O, and S, wherein said C6-10 aryl and 5-10 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RA.
7. The compound of claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl optionally substituted by 1 or 2 substituents independently selected from RA.
8. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl substituted by one RA.
9. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl substituted by CN.
   
10. The compound of any one of claims 1 and 4 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl or 5-6 membered heteroaryl comprising carbon and 1, 2, 3 or 4 heteroatoms selected from N, O, and S; wherein said phenyl and 5-6 membered heteroaryl are each optionally substituted by 1, 2, 3, or 4 substituents independently selected from RB.
11. The compound of any one of claims 1, 2, and 4 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl optionally substituted by 1 or 2 substituents independently selected from RB.
12. The compound of any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl substituted by one RB.
13. The compound of any one of claims 1 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl substituted by methyl.
14. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R1 is halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, Cy1, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, NRc1C(O)ORa1, S(O)2Rb1, or S(O)2NRc1Rd1; wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1, 2, or 3 substituents independently selected from Cy1, halo, CN, ORa1, C(O)Rb1, C(O)NRc1Rd1, C(O)ORa1, NRc1Rd1, NRc1C(O)Rb1, S(O)2Rb1, and S(O)2NRc1Rd1.
15. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R1 is C1-6 alkyl, Cy1, or ORa1, wherein said C1-6 alkyl is substituted with one Cy1.
16. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R1 is pyrrolidin-3-ylmethoxy, 2-pyrrolidin-3-ylethyl, (1-methylpyrrolidin-3- yl)ethyl, 3-[(methylamino)methyl]phenyl, 3-aminopyrrolidin-1-yl)methyl]phenyl, piperazin-1- ylmethyl, 4-methylpiperazin-1-yl)methyl, 3-(dimethylamino)pyrrolidin-1-yl, 3- (methylamino)pyrrolidin-1-yl, or (1-methylpyrrolidin-3-yl)methoxy.    
17. The compound of any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof, wherein R2 is H.
18. The compound of any one of claims 1 and 4 to 17, or a pharmaceutically acceptable salt thereof, wherein each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, ORa4, C(O)Rb4, C(O)NRc4Rd4, C(O)ORa4, NRc4Rd4, NRc4C(O)Rb4, S(O)2Rb4, and S(O)2NRc4Rd4.
19. The compound of any one of claims 1, 2, and 4 to 17, or a pharmaceutically acceptable salt thereof, wherein each RA is independently selected from halo, C1-6 alkyl, C1-6 haloalkyl, CN, and ORa4, wherein said C1-6 alkyl is optionally substituted by 1, 2, or 3, substituents
independently selected from CN and ORa4.
20. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein RA is CN.
21. The compound of any one of claims 1 and 4 to 20, or a pharmaceutically acceptable salt thereof, wherein each RB is independently selected from Cy3, halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5,
NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5, wherein said C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, or 3 substituents independently selected from Cy3, halo, C1-6 haloalkyl, CN, NO2, ORa5, C(O)Rb5, C(O)NRc5Rd5, C(O)ORa5, NRc5Rd5,
NRc5C(O)Rb5, S(O)2Rb5, and S(O)2NRc5Rd5.
22. The compound of any one of claims 1, 2, and 4 to 20, or a pharmaceutically acceptable salt thereof, wherein each RB is independently selected from halo, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, CN, and ORa5.
   
23. The compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt thereof, wherein RB is C1-6 alkyl.
24. The compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt thereof, wherein RB is methyl.
25. The compound of any one of claims 1 to 24, or a pharmaceutically acceptable salt thereof, wherein RX is H.
26. The compound of any one of claims 1, 2, and 4 to 25, or a pharmaceutically acceptable salt thereof, wherein each Cy1 is independently selected from phenyl, C3-7 cycloalkyl, 5-6 membered heteroaryl, and 4-7 membered heterocycloalkyl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from RCy.
27. The compound of any one of claims 1 to 25, or a pharmaceutically acceptable salt thereof, wherein each Cy1 is phenyl or 4-7 membered heterocycloalkyl, each optionally substituted with 1 or 2 substituents independently selected from RCy.
28. The compound of any one of claims 1 to 25, or a pharmaceutically acceptable salt thereof, wherein each Cy1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from RCy.
29. The compound of any one of claims 1 to 25, or a pharmaceutically acceptable salt thereof, wherein each Cy1 is phenyl, pyrrolidinyl, or piperazinyl, each optionally substituted with 1 or 2 substituents independently selected from C1-4 alkyl and NRc6Rd6, wherein said C1-4 alkyl is optionally substituted with NRc6Rd6. 30. The compound of any one of claims 1 to 29, or a pharmaceutically acceptable salt thereof, wherein RCy is C1-4 alkyl and NRc6Rd6, wherein said C1-4 alkyl is optionally substituted with NRc6Rd6.
   
30. The compound of any one of claims 1 and 4 to 30, or a pharmaceutically acceptable salt thereof, wherein each Ra1 is independently selected from H, C1-6 alkyl, and Cy4; wherein said C1- 6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3.
31. The compound of any one of claims 1, 2, and 4 to 30, or a pharmaceutically acceptable salt thereof, wherein each Ra1 is independently selected from H, C1-6 alkyl, and 4-7 membered heterocycloalkyl; wherein said C1-6 alkyl is optionally substituted with 1, 2, or 3 substituents independently selected from Cy4, halo, CN, ORa3, C(O)Rb3, C(O)NRc3Rd3, C(O)ORa3, NRc3Rd3, NRc3C(O)Rb3, S(O)2Rb3, and S(O)2NRc3Rd3, and wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1, 2, or 3 substituents independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, S(O)2Rb6, and S(O)2NRc6Rd6.
32. The compound of any one of claims 1 to 30, or a pharmaceutically acceptable salt thereof, wherein each Ra1 is C1-4 alkyl substituted by 4-7 membered heterocycloalkyl, wherein said 4-7 membered heterocycloalkyl is optionally substituted with 1 or 2 substituents
independently selected from halo, C1-4 alkyl, C1-4 haloalkyl, C1-4 cyanoalkyl, CN, NO2, ORa6, C(O)Rb6, C(O)NRc6Rd6, C(O)ORa6, NRc6Rd6, NRc6C(O)Rb6, S(O)2Rb6, and S(O)2NRc6Rd6.
33. The compound of any one of claims 1 to 30, or a pharmaceutically acceptable salt thereof, wherein each Ra1 is pyrrolidinylmethyl optionally substituted with one C1-4 alkyl.
34. The compound of any one of claims 1 to 3, 6 to 24, and 26 to 33 having Formula IIa:
Figure imgf000089_0001
   
IIa
or a pharmaceutically acceptable salt thereof.
35. The compound of any one of claims 1 to 3 and 6 to 33 having Formula IIb:
Figure imgf000090_0001
or a pharmaceutically acceptable salt thereof.
36. The compound of any one of claims 1 to 3, 14 to 16, and 26 to 33 having Formula IIIa:
Figure imgf000090_0002
or a pharmaceutically acceptable salt thereof.
37. The compound of any one of claims 1 to 3, 14 to 16, and 26 to 33 having Formula IIIb:
Figure imgf000090_0003
or a pharmaceutically acceptable salt thereof.
   
38. The compound of claim 1 selected from:
4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyridin-6- yl}benzonitrile;
4-[5-(4-methylphenyl)-8-(2-pyrrolidin-3-ylethyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile;
4-{5-(4-methylphenyl)-8-[2-(1-methylpyrrolidin-3-yl)ethyl][1,2,4]triazolo[1,5-a]pyridin- 6-yl}benzonitrile;
4-[8-{3-[(methylamino)methyl]phenyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5-a]pyridin- 6-yl]benzonitrile;
4-[8-{3-[(3-aminopyrrolidin-1-yl)methyl]phenyl}-5-(4-methylphenyl)[1,2,4]triazolo[1,5- a]pyridin-6-yl]benzonitrile;
4-[5-(4-methylphenyl)-8-(piperazin-1-ylmethyl)[1,2,4]triazolo[1,5-a]pyridin-6- yl]benzonitrile;
4-{5-(4-methylphenyl)-8-[(4-methylpiperazin-1-yl)methyl][1,2,4]triazolo[1,5-a]pyridin- 6-yl}benzonitrile;
4-[8-{[(3S)-3-(dimethylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)- [1,2,4]triazolo-[1,5-a]pyridin-6-yl]benzonitrile;
4-[8-{[(3R)-3-(dimethylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)- [1,2,4]triazolo-[1,5-a]pyridin-6-yl]benzonitrile;
4-[8-{[(3S)-3-(methylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)- [1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile;
4-[8-{[(3R)-3-(methylamino)pyrrolidin-1-yl]methyl}-5-(4-methylphenyl)- [1,2,4]triazolo[1,5-a]pyridin-6-yl]benzonitrile;
4-{5-(4-methylphenyl)-8-[(3R)-pyrrolidin-3-ylmethoxy][1,2,4]triazolo[1,5-a]pyrazin-6- yl}benzonitrile; and
4-(5-(4-methylphenyl)-8-{[(3R)-1-methylpyrrolidin-3-yl]methoxy}[1,2,4]triazolo[1,5- a]pyrazin-6-yl)benzonitrile,
or a pharmaceutically acceptable salt of any of the aforementioned.
   
39. A pharmaceutical composition comprising a compound of any one of claims 1 to 38, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
40. A method of inhibiting LSD1 comprising contacting a compound of any one of claims 1 to 38, or a pharmaceutically acceptable salt thereof, with said LSD1.
41. A method of treating a disease comprising administering to a patient a therapeutically effective amount of a compound of any one of claims 1 to 38, or a pharmaceutically acceptable salt thereof, wherein said disease is cancer.
42. The method of claim 41 wherein said cancer is a hematological cancer.
43. The method of claim 42 wherein said hematological cancer is selected from acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Non-Hodgkin lymphoma (including relapsed or refractory NHL and recurrent follicular), Hodgkin lymphoma, primary myelofibrosis (PMF), polycythemia vera (PV), essential thrombocytosis (ET)), myelodysplasia syndrome (MDS), or multiple myeloma.
44. The method of claim 41 wherein said cancer is a sarcoma, lung cancer, gastrointestinal cancer, genitourinary tract cancer, liver cancer, bone cancer, nervous system cancer,
gynecological cancer, or skin cancer.
45. A method of treating a disease comprising administering to a patient a therapeutically effective amount of a compound of any one of claims 1 to 38, or a pharmaceutically acceptable salt thereof, wherein said disease is a viral disease or a beta-globinopathy.
   
PCT/US2015/039706 2014-07-10 2015-07-09 Triazolopyridines and triazolopyrazines as lsd1 inhibitors WO2016007722A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462022929P 2014-07-10 2014-07-10
US62/022,929 2014-07-10

Publications (1)

Publication Number Publication Date
WO2016007722A1 true WO2016007722A1 (en) 2016-01-14

Family

ID=53682875

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/039706 WO2016007722A1 (en) 2014-07-10 2015-07-09 Triazolopyridines and triazolopyrazines as lsd1 inhibitors

Country Status (2)

Country Link
US (4) US9695167B2 (en)
WO (1) WO2016007722A1 (en)

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016161282A1 (en) * 2015-04-03 2016-10-06 Incyte Corporation Heterocyclic compounds as lsd1 inhibitors
WO2016198649A1 (en) 2015-06-12 2016-12-15 Oryzon Genomics, S.A. Biomarkers associated with lsd1 inhibitors and uses thereof
WO2017013061A1 (en) 2015-07-17 2017-01-26 Oryzon Genomics, S.A. Biomarkers associated with lsd1 inhibitors and uses thereof
CN106432248A (en) * 2016-09-27 2017-02-22 郑州大学 Pyrimidine and triazole containing LSD1 inhibitor and preparation method and application thereof
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
CN106928235A (en) * 2017-05-03 2017-07-07 郑州大学 The LSD1 of triazole containing pyrimido inhibitor, its preparation method and application
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
WO2017158136A1 (en) 2016-03-16 2017-09-21 Oryzon Genomics, S.A. Methods to determine kdm1a target engagement and chemoprobes useful therefor
WO2018083189A1 (en) 2016-11-03 2018-05-11 Oryzon Genomics, S.A. Biomarkers for determining responsiveness to lsd1 inhibitors
US9994546B2 (en) 2014-02-13 2018-06-12 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
WO2019025588A1 (en) 2017-08-03 2019-02-07 Oryzon Genomics, S.A. Methods of treating behavior alterations
KR20190030192A (en) 2017-09-13 2019-03-21 한미약품 주식회사 Pyrazole derivative compound and use thereof
JP2019508447A (en) * 2016-03-15 2019-03-28 セルジーン クオンティセル リサーチ,インク. Histone demethylase inhibitors
WO2019068326A1 (en) 2017-10-05 2019-04-11 Université D'aix-Marseille Lsd1 inhibitors for the treatment and prevention of cardiomyopathies
KR20190040763A (en) 2017-10-11 2019-04-19 한미약품 주식회사 Pyrazolopyridine derivative compound and use thereof
KR20190040783A (en) 2017-10-11 2019-04-19 한미약품 주식회사 Pyrazole derivatives as Lysine-specific histone demethylase-1 inhibitors
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
US10513493B2 (en) 2014-02-13 2019-12-24 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
WO2020159285A1 (en) 2019-02-01 2020-08-06 한미약품 주식회사 Imidazopyridine derivative compounds and use of same
KR20200096157A (en) 2019-02-01 2020-08-11 한미약품 주식회사 Imidazopyridine derivative compound and use thereof
WO2020188089A1 (en) 2019-03-20 2020-09-24 Oryzon Genomics, S.A. Methods of treating attention deficit hyperactivity disorder using kdm1a inhibitors such as the compound vafidemstat
WO2020188090A1 (en) 2019-03-20 2020-09-24 Oryzon Genomics, S.A. Methods of treating borderline personality disorder
WO2021004610A1 (en) 2019-07-05 2021-01-14 Oryzon Genomics, S.A. Biomarkers and methods for personalized treatment of small cell lung cancer using kdm1a inhibitors
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11161850B2 (en) 2018-07-05 2021-11-02 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
US11168089B2 (en) 2018-05-18 2021-11-09 Incyte Corporation Fused pyrimidine derivatives as A2A / A2B inhibitors
EP3964204A1 (en) 2020-09-08 2022-03-09 Université d'Aix-Marseille Lsd1 inhibitors for use in the treatment and prevention of fibrosis of tissues
US11390624B2 (en) 2019-01-29 2022-07-19 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
WO2022214303A1 (en) 2021-04-08 2022-10-13 Oryzon Genomics, S.A. Combinations of lsd1 inhibitors for treating myeloid cancers
CN115772170A (en) * 2021-12-03 2023-03-10 徐诺药业(南京)有限公司 Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof
US11673894B2 (en) 2018-02-27 2023-06-13 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
WO2023217784A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating nf1-mutant tumors using lsd1 inhibitors
WO2023217758A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating malignant peripheral nerve sheath tumor (mpnst) using lsd1 inhibitors

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3392244A1 (en) 2014-02-13 2018-10-24 Incyte Corporation Cyclopropylamines as lsd1 inhibitors
CN107073006A (en) 2014-09-16 2017-08-18 赛尔基因昆蒂赛尔研究公司 Histone demethylase inhibitors
US9896436B2 (en) 2014-09-16 2018-02-20 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
JP7352284B2 (en) 2017-05-15 2023-09-28 ザ・リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミシガン Pyrrolo[2,3-c]pyridine and related analogs as LSD-1 inhibitors
WO2019083971A1 (en) 2017-10-23 2019-05-02 Children's Medical Center Corporation Methods of treating cancer using lsd1 inhibitors in combination with immunotherapy
US11944614B2 (en) 2018-05-15 2024-04-02 Regents Of The University Of Michigan Imidazo[4,5-c]pyridine compounds as LSD-1 inhibitors
GB202115017D0 (en) 2021-10-20 2021-12-01 Univ London Queen Mary Sequential treatments and biomarkers to reverse resistance to kinase inhibitors
WO2023067058A1 (en) 2021-10-20 2023-04-27 Queen Mary University Of London Sequential treatments and biomarkers to reverse resistance to kinase inhibitors

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2013131609A1 (en) * 2012-03-07 2013-09-12 Merck Patent Gmbh Triazolopyrazine derivatives

Family Cites Families (312)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL7013068A (en) 1969-09-17 1971-03-19
US4537889A (en) 1982-12-27 1985-08-27 Eli Lilly And Company Inotropic agents
US4614810A (en) 1984-09-24 1986-09-30 Pennwalt Corporation 4,5-dihydro-4-oxo-2-[(2-trans-phenylcyclopropyl)amino]-3-furancarboxylic acids and derivatives thereof
US4625040A (en) 1984-09-24 1986-11-25 Pennwalt Corporation N-(phenyl) or N-(phenylcyclopropyl)-2,5-dihydro-2-oxo-4[(substituted phenyl)amino]-3-furancarboxamide derivatives
FR2607813B1 (en) 1986-12-05 1989-03-31 Montpellier I Universite ALKYLAMINO-8 IMIDAZO (1,2-A) PYRAZINES AND DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
JPH032778Y2 (en) 1986-12-15 1991-01-24
AU622330B2 (en) 1989-06-23 1992-04-02 Takeda Chemical Industries Ltd. Condensed heterocyclic compounds having a nitrogen atom in the bridgehead for use as fungicides
JP2844351B2 (en) 1989-07-13 1999-01-06 株式会社科薬 Aqueous solution of stable polymyxin antibiotics
IL96432A0 (en) 1989-11-30 1991-08-16 Schering Ag Pesticidal compositions containing pyridine derivatives and novel pyridine derivatives
FR2662163A1 (en) 1990-05-16 1991-11-22 Lipha New 8-amino-1,2,4-triazolo[4,3-a]pyrazines, preparation processes and medicaments containing them
ES2130269T3 (en) 1992-06-17 1999-07-01 Upjohn Co SUBSTITUTED OXIMES WITH USEFUL PIRIDINE, PIRROLIDINE AND AZEPINE AS AGENTS AGAINST ATHEROSCLEROSIS AND ANTI-HYPERCHOLESTEROLEMICS.
DE4327027A1 (en) 1993-02-15 1994-08-18 Bayer Ag Imidazoazine
FR2711993B1 (en) 1993-11-05 1995-12-01 Rhone Poulenc Rorer Sa Drugs containing 7H-imidazol [1,2-a] pyrazine-8-one derivatives, new compounds and their preparation.
US5932223A (en) 1996-09-26 1999-08-03 Merck & Co., Inc. Rotavirus vaccine formulations
BR9814628A (en) 1997-11-11 2001-11-27 Ono Pharmaceutical Co Fused pyrazine derivatives
JP2000319278A (en) 1999-05-11 2000-11-21 Ono Pharmaceut Co Ltd Condensed pyrazine compound and medicinal agent having the same as active ingredient
JP2000319277A (en) 1999-05-11 2000-11-21 Ono Pharmaceut Co Ltd Condensed pyrazine compound and medicinal agent having the same as active ingredient
JP4032566B2 (en) 1999-06-21 2008-01-16 東レ株式会社 Light emitting element
JP4041624B2 (en) 1999-07-21 2008-01-30 三井化学株式会社 Organic electroluminescence device
JP2001057292A (en) 1999-08-20 2001-02-27 Toray Ind Inc Luminescent element
UA80667C2 (en) 1999-09-28 2007-10-25 Panacea Biotec Ltd Controlled release composition comprising nimesulide and process for the preparation thereof
SE9903611D0 (en) 1999-10-06 1999-10-06 Astra Ab Novel compounds III
DE19948434A1 (en) 1999-10-08 2001-06-07 Gruenenthal Gmbh Substance library containing bicyclic imidazo-5-amines and / or bicyclic imidazo-3-amines
JP4409680B2 (en) 1999-10-18 2010-02-03 株式会社ヤクルト本社 Tricyclic fused imidazole derivatives
US6403588B1 (en) 2000-04-27 2002-06-11 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
CA2407573C (en) 2000-04-27 2011-09-13 Yamanouchi Pharmaceutical Co. Ltd. Imidazopyridine derivatives
AR029538A1 (en) 2000-07-06 2003-07-02 Wyeth Corp PHARMACEUTICAL COMPOSITIONS OF ESTROGEN AGENTS
US6589952B2 (en) 2000-07-14 2003-07-08 Bristol-Myers Squibb Pharma Company Imidazo[1,2-a]pyrazines for the treatment of neurological disorders
DE10050663A1 (en) 2000-10-13 2002-04-18 Gruenenthal Gmbh Use of amino-substituted imidazo(1,2-a)pyridine, imidazo(1,2-a)pyrimidine and imidazo(1,2-a)pyrazine derivatives as NO synthase inhibitors, e.g. in treatment of migraine and neurodegenerative diseases
WO2002034748A1 (en) 2000-10-24 2002-05-02 Sankyo Company, Limited Imidazopyridine derivatives
JP2002205992A (en) 2000-11-08 2002-07-23 Takeda Chem Ind Ltd Bicyclic triazolone derivative and herbicide comprising the same
ATE310740T1 (en) 2000-11-10 2005-12-15 Merck Sharp & Dohme IMIDAZOTRIAZINE DERIVATIVES AS LIGANDS FOR GABA RECEPTORS
EP1343785A2 (en) 2000-12-13 2003-09-17 Basf Aktiengesellschaft Use of substituted imidazoazines, novel imidazoazines, methods for the production thereof, and agents containing these compounds
EP1217000A1 (en) 2000-12-23 2002-06-26 Aventis Pharma Deutschland GmbH Inhibitors of factor Xa and factor VIIa
TWI312347B (en) 2001-02-08 2009-07-21 Eisai R&D Man Co Ltd Bicyclic nitrogen-containing condensed ring compounds
WO2002072549A1 (en) 2001-03-12 2002-09-19 Millennium Pharmaceuticals, Inc. Functionalized heterocycles as modulators of chemokine receptor function and methods of use therefor
AR035543A1 (en) 2001-06-26 2004-06-16 Japan Tobacco Inc THERAPEUTIC AGENT FOR HEPATITIS C THAT INCLUDES A CONDENSED RING COMPOUND, CONDENSED RING COMPOUND, PHARMACEUTICAL COMPOSITION THAT UNDERSTANDS, BENZIMIDAZOL, THIAZOL AND BIFENYL COMPOUNDS USED AS INTERMEDIARY COMPARTMENTS OF COMPARTMENTS
IL159811A0 (en) 2001-07-13 2004-06-20 Neurogen Corp Heteroaryl substituted fused bicyclic heteroaryl compounds as gabaa receptor ligands
US6921762B2 (en) 2001-11-16 2005-07-26 Amgen Inc. Substituted indolizine-like compounds and methods of use
JP2005519915A (en) 2002-01-18 2005-07-07 セレテック・リミテッド・ライアビリティ・カンパニー Method for treating symptoms associated with Edg receptors
US20050113283A1 (en) 2002-01-18 2005-05-26 David Solow-Cordero Methods of treating conditions associated with an EDG-4 receptor
WO2004017950A2 (en) 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents
UA80296C2 (en) 2002-09-06 2007-09-10 Biogen Inc Imidazolopyridines and methods of making and using the same
US20060276339A1 (en) 2002-10-16 2006-12-07 Windsor J B Methods and compositions for increasing the efficacy of biologically-active ingredients
BR0317430A (en) 2002-12-20 2005-10-25 Pharmacia Corp Mitogen-activated protein kinase-activated protein kinase-2 inhibitor compounds
US7189723B2 (en) 2003-02-10 2007-03-13 Cgi Pharmaceuticals, Inc. Certain 8-heteroaryl-6-phenyl-imidazo[1,2-a]pyrazines as modulators of kinase activity
US7157460B2 (en) 2003-02-20 2007-01-02 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
US7186832B2 (en) 2003-02-20 2007-03-06 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
GB0303910D0 (en) 2003-02-20 2003-03-26 Merck Sharp & Dohme Therapeutic agents
EP1604981A4 (en) 2003-03-14 2008-12-24 Ono Pharmaceutical Co Nitrogen-containing heterocyclic derivatives and drugs containing the same as the active ingredient
WO2004089416A2 (en) 2003-04-11 2004-10-21 Novo Nordisk A/S Combination of an 11beta-hydroxysteroid dehydrogenase type 1 inhibitor and an antihypertensive agent
ATE455547T1 (en) 2003-04-11 2010-02-15 High Point Pharmaceuticals Llc PHARMACEUTICAL USES OF CONDENSED 1,2,4-TRIAZOLES
ATE512957T1 (en) 2003-04-24 2011-07-15 Merck Sharp & Dohme ACT ACTIVITY INHIBITOR
SE0301653D0 (en) 2003-06-05 2003-06-05 Astrazeneca Ab Novel compounds
MXPA06000554A (en) 2003-07-14 2006-07-03 Arena Pharm Inc Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto.
US7538120B2 (en) 2003-09-03 2009-05-26 Array Biopharma Inc. Method of treating inflammatory diseases
WO2005025558A1 (en) 2003-09-12 2005-03-24 Applied Reserach Systems Ars Holding N.V. Sulfonamide derivatives for the treatment of diabetes
JP2005089352A (en) 2003-09-16 2005-04-07 Kissei Pharmaceut Co Ltd NEW IMIDAZO[1,5-a]PYRAZINE DERIVATIVE, MEDICINE COMPOSITION CONTAINING THE SAME AND THEIR USE
ATE369370T1 (en) 2003-10-10 2007-08-15 Pfizer Prod Inc SUBSTITUTED 2H-(1,2,4)TRIAZOLO(4,3-A)PYRAZINE AS GSK-3 INHIBITORS
US7419978B2 (en) 2003-10-22 2008-09-02 Bristol-Myers Squibb Company Phenyl-aniline substituted bicyclic compounds useful as kinase inhibitors
WO2005044793A2 (en) 2003-10-31 2005-05-19 Takeda Pharmaceutical Company Limited Nitrogen-containing fused heterocyclic compounds
WO2005063241A1 (en) 2003-12-26 2005-07-14 Ono Pharmaceutical Co., Ltd. Preventive and/or therapeutic agent for disease in which mitochondrial benzodiazepine receptor participates
US20070191395A1 (en) * 2004-02-16 2007-08-16 Katsuhiro Kawakami Heterocyclic compounds having antifungal activity
US7306631B2 (en) 2004-03-30 2007-12-11 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
EP1758557B1 (en) 2004-05-11 2011-07-13 Egalet Ltd. Swellable dosage form comprising gellan gum
WO2006015263A2 (en) 2004-07-29 2006-02-09 Threshold Pharmaceuticals, Inc. Lonidamine analogs
MX2007001612A (en) 2004-08-18 2007-04-10 Upjohn Co Triazolopyridine compounds useful for the treatment of inflammation.
WO2006026703A2 (en) 2004-09-02 2006-03-09 Smithkline Beecham Corporation Chemical compounds
BRPI0516819A (en) 2004-10-07 2008-09-23 Warner Lambert Co triazolopyridine derivatives and antibacterial agents
WO2006057946A2 (en) 2004-11-22 2006-06-01 Threshold Pharmaceuticals, Inc. Tubulin binding anti cancer agents and prodrugs thereof
AU2005311451A1 (en) 2004-12-01 2006-06-08 Merck Serono Sa [1,2,4]triazolo[4,3-a]pyridine derivatives for the treatment of hyperproliferative diseases
US20070293456A9 (en) 2004-12-30 2007-12-20 Anthony Hayford Method for the synthesis of 3-substituted indolizine and benzoindolizine compounds
US7456289B2 (en) 2004-12-31 2008-11-25 National Health Research Institutes Anti-tumor compounds
EA012615B1 (en) 2005-02-22 2009-10-30 Пфайзер Инк. Oxyindole derivatives as 5htreceptor agonists
ITBO20050123A1 (en) 2005-03-07 2005-06-06 Alfa Wassermann Spa GASTRORESISTIC PHARMACEUTICAL FORMULATIONS CONTAINING RIFAXIMINA
AR053712A1 (en) 2005-04-18 2007-05-16 Neurogen Corp HETEROARILOS SUBSTITUTED, ANTAGONISTS OF CB1 (RECEIVER 1 CANABINOID)
EP1901748B1 (en) 2005-06-09 2010-09-08 Oncalis AG Angiogenesis inhibitors
US7572807B2 (en) 2005-06-09 2009-08-11 Bristol-Myers Squibb Company Heteroaryl 11-beta-hydroxysteroid dehydrogenase type I inhibitors
US7579360B2 (en) 2005-06-09 2009-08-25 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
US7632837B2 (en) 2005-06-17 2009-12-15 Bristol-Myers Squibb Company Bicyclic heterocycles as cannabinoid-1 receptor modulators
TW200726765A (en) 2005-06-17 2007-07-16 Bristol Myers Squibb Co Triazolopyridine cannabinoid receptor 1 antagonists
US7452892B2 (en) 2005-06-17 2008-11-18 Bristol-Myers Squibb Company Triazolopyrimidine cannabinoid receptor 1 antagonists
WO2007028051A2 (en) 2005-09-02 2007-03-08 Abbott Laboratories Novel imidazo based heterocycles
JP2009507843A (en) 2005-09-09 2009-02-26 シェーリング コーポレイション Aza-fused cyclin-dependent kinase inhibitors
JP5031760B2 (en) 2005-11-10 2012-09-26 シェーリング コーポレイション Imidazopyrazine as a protein kinase inhibitor
JP5334588B2 (en) 2005-11-28 2013-11-06 マリナス ファーマシューティカルズ Ganaxolone preparation, method for producing the same, and use thereof
ATE537171T1 (en) 2005-12-27 2011-12-15 Hoffmann La Roche ARYL-ISOXAZOLE-4-YL-IMIDAZOÄ1,5-AÜPYRIDINE DERIVA E
WO2007074491A1 (en) 2005-12-28 2007-07-05 Universita Degli Studi Di Siena HETEROTRICYCLIC AMIDE DERIVATIVES AS NEUROKININ-l (NKl) RECEPTOR LIGANDS
PE20070978A1 (en) 2006-02-14 2007-11-15 Novartis Ag HETEROCICLIC COMPOUNDS AS INHIBITORS OF PHOSPHATIDYLINOSITOL 3-KINASES (PI3Ks)
US8097617B2 (en) 2006-03-31 2012-01-17 Novartis Ag Organic compounds
AR061229A1 (en) 2006-06-06 2008-08-13 Schering Corp IMIDAZOPIRAZINS AS INHIBITORS OF PROTEIN QUINASA
US20090175852A1 (en) 2006-06-06 2009-07-09 Schering Corporation Imidazopyrazines as protein kinase inhibitors
AU2007261397A1 (en) 2006-06-22 2007-12-27 Mallinckrodt Llc Pyrazine derivatives with extended conjugation and uses thereof
JP2009534396A (en) 2006-06-22 2009-09-24 マリンクロッド・インコーポレイテッド Pyrazine derivatives and their use in kidney monitoring
CN101506198A (en) 2006-06-29 2009-08-12 先灵公司 Substituted bicyclic and tricyclic thrombin receptor antagonists
WO2008005423A1 (en) 2006-07-03 2008-01-10 Cambrex Charles City, Inc. Improved method of making sufentanil
WO2008005908A2 (en) 2006-07-07 2008-01-10 Forest Laboratories Holdings Limited Pyridoimidazole derivatives
US8198448B2 (en) 2006-07-14 2012-06-12 Amgen Inc. Fused heterocyclic derivatives and methods of use
PE20080403A1 (en) 2006-07-14 2008-04-25 Amgen Inc FUSED HETEROCYCLIC DERIVATIVES AND METHODS OF USE
US8217177B2 (en) 2006-07-14 2012-07-10 Amgen Inc. Fused heterocyclic derivatives and methods of use
US20080021217A1 (en) 2006-07-20 2008-01-24 Allen Borchardt Heterocyclic inhibitors of rho kinase
WO2008027812A2 (en) 2006-08-28 2008-03-06 Forest Laboratories Holdings Limited Imidazopyridine and imidazopyrimidine derivatives
DE102006041292A1 (en) 2006-09-01 2008-03-06 Henkel Kgaa Use of optionally substituted hexagonal heterocycle with a nitrogen in the ring for activating and improving the brightening effect of the hydrogen peroxide for keratin fibers such as fur, wool, feathers and human hair
WO2008037607A1 (en) 2006-09-25 2008-04-03 Basf Se Heterocyclic compounds containing carbonyl groups, and the use thereof for controlling phytopathogenic fungi
CA2665195C (en) 2006-10-11 2011-08-09 Amgen Inc. Imidazo- and triazolo-pyridine compounds and methods of use thereof
CA2669094A1 (en) 2006-11-08 2008-05-29 Novavax,Inc. Method of preparing solid dosage forms of multi-phasic pharmaceutical compositions
WO2008056176A1 (en) 2006-11-10 2008-05-15 Scottish Biomedical Limited Pyrazolopyrimidines as phosphodiesterase inhibitors
NO346024B1 (en) 2006-11-22 2022-01-03 Incyte Holdings Corp IMIDAZOTRIAZINES AND IMIDAZOPYRIMIDINES AS CHINESE INHIBITORS
EP2086540B8 (en) 2006-12-01 2011-03-02 Galapagos N.V. Triazolopyridine compounds useful for the treatment of degenerative & inflammatory diseases
CA2673511A1 (en) 2006-12-22 2008-07-03 Combinatorx, Incorporated Pharmaceutical compositions for treatment of parkinson's disease and related disorders
EP2118101B1 (en) 2007-03-09 2012-09-26 Probiodrug AG Imidazo [1,5-a] pyridine derivatives as inhibitors of glutaminyl cyclase
DE102007012645A1 (en) 2007-03-16 2008-09-18 Bayer Healthcare Ag Substituted imidazo and triazolopyrimidines
EP1972628A1 (en) 2007-03-21 2008-09-24 Schwarz Pharma Ag Indolizines and aza-analog derivatives thereof as CNS active compounds
JP2010523725A (en) 2007-04-16 2010-07-15 レオ ファーマ アクティーゼルスカブ Triazolopyridines as phosphodiesterase inhibitors for the treatment of skin diseases
WO2008130951A1 (en) 2007-04-17 2008-10-30 Bristol-Myers Squibb Company Fused heterocyclic 11-beta-hydroxysteroid dehydrogenase type i inhibitors
US20110206607A1 (en) 2007-05-10 2011-08-25 Roger Olsson Imidazol (1,2-a)pyridines and related compounds with activity at cannabinoid cb2 receptors
JP5343845B2 (en) 2007-05-21 2013-11-13 東レ株式会社 Oral preparation containing specific organic acid and method for improving dissolution and chemical stability of oral preparation
EP2167491A1 (en) 2007-06-08 2010-03-31 Abbott Laboratories 5-heteroaryl substituted indazoles as kinase inhibitors
US8648069B2 (en) 2007-06-08 2014-02-11 Abbvie Inc. 5-substituted indazoles as kinase inhibitors
JP2010529195A (en) 2007-06-14 2010-08-26 シェーリング コーポレイション Imidazopyrazine as an inhibitor of protein kinase
CL2008001839A1 (en) 2007-06-21 2009-01-16 Incyte Holdings Corp Compounds derived from 2,7-diazaspirocycles, inhibitors of 11-beta hydroxyl steroid dehydrogenase type 1; pharmaceutical composition comprising said compounds; Useful to treat obesity, diabetes, glucose intolerance, type II diabetes, among other diseases.
US20090004281A1 (en) 2007-06-26 2009-01-01 Biovail Laboratories International S.R.L. Multiparticulate osmotic delivery system
US8053574B2 (en) 2007-07-18 2011-11-08 Novartis Ag Organic compounds
CN101808666A (en) 2007-07-31 2010-08-18 先灵公司 Anti-mitotic agent and aurora kinase inhibitor combination as anti-cancer treatment
WO2009017954A1 (en) 2007-08-01 2009-02-05 Phenomix Corporation Inhibitors of jak2 kinase
JP2010535773A (en) 2007-08-10 2010-11-25 グラクソスミスクライン エルエルシー Nitrogen-containing bicyclic chemicals for treating viral infections
US20090047336A1 (en) 2007-08-17 2009-02-19 Hong Kong Baptist University novel formulation of dehydrated lipid vesicles for controlled release of active pharmaceutical ingredient via inhalation
FR2920090A1 (en) 2007-08-24 2009-02-27 Oreal Composition for coloring keratin fibers, preferably human hair, comprises amino pyrazolopyridine oxidation bases, couplers, and surfactants comprising alkyl ether carboxylic acid and alkyl polyglucosides
FR2920091A1 (en) 2007-08-24 2009-02-27 Oreal Composition for coloring keratin fibers, preferably human hair, comprises amino pyrazolopyridine oxidation bases, couplers and polyols comprising hydrocarbon chain carrying two hydroxyl functions, where the chain is free from ether function
KR20090022616A (en) 2007-08-31 2009-03-04 한올제약주식회사 Oral administration drug, which contains clopidogrel besylate
US8119658B2 (en) 2007-10-01 2012-02-21 Bristol-Myers Squibb Company Triazolopyridine 11-beta hydroxysteroid dehydrogenase type I inhibitors
GB0719803D0 (en) 2007-10-10 2007-11-21 Cancer Rec Tech Ltd Therapeutic compounds and their use
NZ585261A (en) 2007-10-11 2011-10-28 Astrazeneca Ab Pyrrolo [2, 3 -d] pyrimidin derivatives as protein kinase b inhibitors
EP3733162A1 (en) 2007-10-12 2020-11-04 Novartis AG Compositions comprising sphingosine 1 phosphate (s1p) receptor modulators
WO2009085230A1 (en) 2007-12-19 2009-07-09 Amgen Inc. Inhibitors of pi3 kinase
AU2008343062B2 (en) 2007-12-19 2013-03-07 Genentech, Inc. 8-Anilinoimidazopyridines and their use as anti-cancer and/or anti-inflammatory agents
KR100988233B1 (en) 2007-12-26 2010-10-18 한미홀딩스 주식회사 Pharmaceutical composition and formulation comprising clopidogrel 1,5-naphthalene disulfonate or hydrate thereof
AU2009223640B2 (en) 2008-03-11 2013-07-04 Incyte Holdings Corporation Azetidine and cyclobutane derivatives as jak inhibitors
WO2009114180A1 (en) 2008-03-13 2009-09-17 The General Hospital Corporation Inhibitors of the bmp signaling pathway
US8637542B2 (en) 2008-03-14 2014-01-28 Intellikine, Inc. Kinase inhibitors and methods of use
EP2277881A4 (en) 2008-04-18 2011-09-07 Shionogi & Co Heterocyclic compound having inhibitory activity on p13k
DE102008023801A1 (en) 2008-05-15 2009-11-19 Bayer Schering Pharma Aktiengesellschaft Substituted imidazo and triazolopyrimidines, imidazo and pyrazolopyrazines and imidazotriazines
US8349210B2 (en) 2008-06-27 2013-01-08 Transitions Optical, Inc. Mesogenic stabilizers
WO2010010188A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases.
WO2010010189A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010187A1 (en) 2008-07-25 2010-01-28 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
WO2010010184A1 (en) 2008-07-25 2010-01-28 Galapagos Nv [1, 2, 4] triazolo [1, 5-a] pyridines as jak inhibitors
UY32049A (en) 2008-08-14 2010-03-26 Takeda Pharmaceutical CMET INHIBITORS
TR200806298A2 (en) 2008-08-22 2010-03-22 Bi̇lgi̇ç Mahmut Pharmaceutical formulation
US20120021519A1 (en) 2008-09-19 2012-01-26 Presidents And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
JP2010070503A (en) 2008-09-19 2010-04-02 Daiichi Sankyo Co Ltd Antifungal 2-amino-triazolopyridine derivative
US8703778B2 (en) 2008-09-26 2014-04-22 Intellikine Llc Heterocyclic kinase inhibitors
WO2010043721A1 (en) 2008-10-17 2010-04-22 Oryzon Genomics, S.A. Oxidase inhibitors and their use
WO2010048149A2 (en) 2008-10-20 2010-04-29 Kalypsys, Inc. Heterocyclic modulators of gpr119 for treatment of disease
ES2403633T3 (en) 2008-12-04 2013-05-20 Proximagen Limited Imidazopyridine Compounds
US8450321B2 (en) 2008-12-08 2013-05-28 Gilead Connecticut, Inc. 6-(1H-indazol-6-yl)-N-[4-(morpholin-4-yl)phenyl]imidazo-[1,2-A]pyrazin-8-amine, or a pharmaceutically acceptable salt thereof, as a SYK inhibitor
EP2389362B1 (en) 2009-01-21 2019-12-11 Oryzon Genomics, S.A. Phenylcyclopropylamine derivatives and their medical use
CA2750517A1 (en) 2009-02-04 2010-08-12 Vitae Pharmaceuticals, Inc. Cyclic inhibitors of 11beta-hydroxysteroid dehydrogenase 1
US20100209489A1 (en) 2009-02-04 2010-08-19 Supernus Pharmaceuticals, Inc. Formulations of desvenlafaxine
TR200900878A2 (en) 2009-02-05 2010-08-23 Bi̇lgi̇ç Mahmut Pharmaceutical formulations combined in a single dosage form
TR200900879A2 (en) 2009-02-05 2010-08-23 Bi̇lgi̇ç Mahmut Pharmaceutical compositions in which the active ingredients are combined in a single dosage form
EP2396331B1 (en) 2009-02-13 2013-10-16 Bayer Intellectual Property GmbH Fused pyrimidines as akt inhibitors
KR20100101054A (en) 2009-03-07 2010-09-16 주식회사 메디젠텍 Composition for treating or preventing nuclear export of gsk3- mediated disease including compound for inhibiting nuclear export of gsk3
WO2010107404A1 (en) 2009-03-16 2010-09-23 Mahmut Bilgic Stable pharmaceutical combinations
WO2010108059A1 (en) 2009-03-20 2010-09-23 Incyte Corporation Substituted pyrimidine derivatives as antagonists of the histamine h4 receptor
CA2755132C (en) 2009-03-31 2018-02-13 Kissei Pharmaceutical Co., Ltd. Indolizine derivative and use thereof for medical purposes
BRPI1014572B8 (en) 2009-04-16 2022-07-19 Fundacion Centro Nac De Investigaciones Oncologicas Carlos Iii IMIDAZOPYRAZINES FOR USE AS KINASE INHIBITORS
TWI461426B (en) 2009-05-27 2014-11-21 Merck Sharp & Dohme (dihydro)imidazoiso[5,1-a]quinolines
AU2010258785A1 (en) 2009-06-10 2012-01-19 Sunovion Pharmaceuticals Inc. Histamine H3 inverse agonists and antagonists and methods of use thereof
CA2766033C (en) 2009-06-25 2016-09-20 Alkermes, Inc. Prodrugs of nh-acidic compounds
PT2467141T (en) 2009-08-17 2019-02-06 Intellikine Llc Heterocyclic compounds and uses thereof
US9708255B2 (en) 2009-08-18 2017-07-18 Robert A. Casero (bis)urea and (bis)thiourea compounds as epigenic modulators of lysine-specific demethylase 1 and methods of treating disorders
EP2473495A1 (en) 2009-09-18 2012-07-11 Almac Discovery Limited Pharmaceutical compounds
WO2011035941A1 (en) 2009-09-25 2011-03-31 Oryzon Genomics S.A. Lysine specific demethylase-1 inhibitors and their use
EP2486002B1 (en) 2009-10-09 2019-03-27 Oryzon Genomics, S.A. Substituted heteroaryl- and aryl- cyclopropylamine acetamides and their use
WO2011050245A1 (en) 2009-10-23 2011-04-28 Yangbo Feng Bicyclic heteroaryls as kinase inhibitors
US8541404B2 (en) 2009-11-09 2013-09-24 Elexopharm Gmbh Inhibitors of the human aldosterone synthase CYP11B2
US8614315B2 (en) 2009-12-25 2013-12-24 Mahmut Bilgic Cefdinir and cefixime formulations and uses thereof
EP2526102B1 (en) 2010-01-22 2017-03-08 Fundación Centro Nacional de Investigaciones Oncológicas Carlos III Inhibitors of PI3 kinase
US20130085133A1 (en) 2010-02-08 2013-04-04 Sourthern Research Institute Office of Commercialization and Intellectual Prop. Anti-viral treatment and assay to screenfor anti-viral agent
US9616058B2 (en) 2010-02-24 2017-04-11 Oryzon Genomics, S.A. Potent selective LSD1 inhibitors and dual LSD1/MAO-B inhibitors for antiviral use
WO2011106106A2 (en) 2010-02-24 2011-09-01 Oryzon Genomics, S.A. Lysine demethylase inhibitors for diseases and disorders associated with hepadnaviridae
TW201200518A (en) 2010-03-10 2012-01-01 Kalypsys Inc Heterocyclic inhibitors of histamine receptors for the treatment of disease
US9468642B2 (en) 2010-03-18 2016-10-18 Bayer Intellectual Property Gmbh Imidazopyrazines
CA2793086C (en) 2010-03-18 2018-08-21 Institut Pasteur Korea Substituted imidazo[1,2-a]pyridine compounds and their use in the treatment of bacterial infections
LT2552920T (en) 2010-04-02 2017-06-12 Ogeda Sa Novel nk-3 receptor selective antagonist compounds, pharmaceutical composition and methods for use in nk-3 receptors mediated disorders
CN102947265B (en) 2010-04-19 2015-07-29 奥瑞泽恩基因组学股份有限公司 Methionin specific demethylase-1 inhibitor and application thereof
ES2564352T3 (en) 2010-04-20 2016-03-22 Università Degli Studi Di Roma "La Sapienza" Tranylcypromine derivatives as histone demethylase inhibitors LSD1 and / or LSD2
MX2012012145A (en) 2010-04-28 2012-11-21 Daiichi Sankyo Co Ltd [5,6] heterocyclic compound.
US20130131057A1 (en) 2010-05-13 2013-05-23 Centro Nacional De Investigaciones Oncologicas (Cnio New bicyclic compounds as pi3-k and mtor inhibitors
SG185515A1 (en) 2010-05-13 2012-12-28 Amgen Inc Nitrogen heterocyclic compounds useful as pde10 inhibitors
CN102247321A (en) 2010-05-20 2011-11-23 上海亚盛医药科技有限公司 Apogossypolone self-emulsifying drug delivery system and preparation method thereof
WO2011149438A1 (en) 2010-05-28 2011-12-01 Mahmut Bilgic Combination of antihypertensive agents
CN102295642B (en) 2010-06-25 2016-04-06 中国人民解放军军事医学科学院毒物药物研究所 2-Aryimidazole is [1,2-a] pyridine-3-acetamide, Preparation Method And The Use also
JP5858586B2 (en) 2010-07-02 2016-02-10 ギリアード サイエンシーズ, インコーポレイテッド Fused heterocyclic compounds as ion channel modulators
CA2804845A1 (en) 2010-07-12 2012-01-19 Stuart Ince Substituted imidazo[1,2-a]pyrimidines and -pyridines
WO2012009475A1 (en) 2010-07-14 2012-01-19 Oregon Health & Science University Methods of treating cancer with inhibition of lysine-specific demethylase 1
CN101987082B (en) 2010-07-16 2013-04-03 钟术光 Solid preparation and preparation method thereof
CN101987081B (en) 2010-07-16 2012-08-08 钟术光 Controlled release preparation
CN103124724B (en) 2010-07-29 2015-05-20 奥瑞泽恩基因组学股份有限公司 Arylcyclopropylamine based demethylase inhibitors of LSD1 and their medical use
US9006449B2 (en) 2010-07-29 2015-04-14 Oryzon Genomics, S.A. Cyclopropylamine derivatives useful as LSD1 inhibitors
WO2012016133A2 (en) 2010-07-29 2012-02-02 President And Fellows Of Harvard College Ros1 kinase inhibitors for the treatment of glioblastoma and other p53-deficient cancers
WO2012034116A2 (en) 2010-09-10 2012-03-15 The Johns Hopkins University Small molecules as epigenetic modulators of lysine-specific demethylase 1 and methods of treating disorders
CN102397552B (en) 2010-09-10 2016-06-08 广州自远生物科技有限公司 A kind of medicine compound preparation of quinolone containing class and its preparation method and application
EP2623505B1 (en) 2010-09-29 2015-07-29 Kissei Pharmaceutical Co., Ltd. (aza)indolizine derivatives as xanthine oxidase inhibitors
US20130303545A1 (en) 2010-09-30 2013-11-14 Tamara Maes Cyclopropylamine derivatives useful as lsd1 inhibitors
US20120108500A1 (en) 2010-10-07 2012-05-03 Naoki Sakane Compositions and Methods for Modulating Immunodeficiency Virus Transcription
CN103188934B (en) 2010-10-18 2015-08-26 纳幕尔杜邦公司 nematocidal sulfonamides
WO2012052745A1 (en) 2010-10-21 2012-04-26 Centro Nacional De Investigaciones Oncológicas (Cnio) Combinations of pi3k inhibitors with a second anti -tumor agent
SI2630146T1 (en) 2010-10-21 2020-12-31 Medivation Technologies Llc Crystaline (8s,9r)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1h-1,2,4-triazol-5-yl)-8,9-dihydro-2h-pyrido(4,3,2-de)phtalazin-3(7h)-one tosylate salt
EP2444084A1 (en) 2010-10-21 2012-04-25 Centro Nacional de Investigaciones Oncológicas (CNIO) Use of PI3K inibitors for the treatment of obesity
WO2012071469A2 (en) 2010-11-23 2012-05-31 Nevada Cancer Institute Histone demethylase inhibitors and uses thereof for treatment o f cancer
WO2012072713A2 (en) 2010-11-30 2012-06-07 Oryzon Genomics, S.A. Lysine demethylase inhibitors for diseases and disorders associated with flaviviridae
EP2651405A2 (en) 2010-12-14 2013-10-23 Electrophoretics Limited Casein kinase 1 (ck1 ) inhibitors
ES2544609T3 (en) 2010-12-17 2015-09-02 Bayer Intellectual Property Gmbh 2-substituted Imidazopyrazines for use as Mps-1 and TTK inhibitors in the treatment of hyper-proliferative disorders
CA2821817A1 (en) 2010-12-17 2012-06-21 Bayer Intellectual Property Gmbh Substituted 6-imidazopyrazines for use as mps-1 and tkk inhibitors in the treatment of hyperproliferative disorders
WO2012080236A1 (en) 2010-12-17 2012-06-21 Bayer Pharma Aktiengesellschaft 6-substituted imidazopyrazines for use as mps-1 and tkk inhibitors in the treatment of hyperproliferative disorders
US20140187548A1 (en) 2010-12-17 2014-07-03 Bayer Intellectual Property Gmbh 6 substituted imidazopyrazines for use as mps-1 and tkk inhibitors in the treatment of hyperproliferative disorders
UY33805A (en) 2010-12-17 2012-07-31 Boehringer Ingelheim Int ? Dihydrobenzofuranyl-piperidinyl, aza-dihydrobenzofuranylpiperidinyl and diaza-dihydrobenzofuranyl-piperidinyl derivatives, pharmaceutical compositions containing them and uses thereof?
EP2655334B1 (en) 2010-12-22 2018-10-03 Eutropics Pharmaceuticals, Inc. Compositions and methods useful for treating diseases
TWI617559B (en) 2010-12-22 2018-03-11 江蘇恆瑞醫藥股份有限公司 2-arylimidazo[1,2-b]pyridazine, 2-phenylimidazo[1,2-a]pyridine, and 2-phenylimidazo[1,2-a]pyrazine derivatives
JP2014504604A (en) 2011-01-21 2014-02-24 ザ ジェネラル ホスピタル コーポレイション Compositions and methods for cardiovascular disease
EP2712316A1 (en) 2011-02-08 2014-04-02 Oryzon Genomics, S.A. Lysine demethylase inhibitors for myeloproliferative or lymphoproliferative diseases or disorders
WO2012107498A1 (en) 2011-02-08 2012-08-16 Oryzon Genomics S.A. Lysine demethylase inhibitors for myeloproliferative disorders
WO2012116237A2 (en) 2011-02-23 2012-08-30 Intellikine, Llc Heterocyclic compounds and uses thereof
BRPI1101013A2 (en) 2011-03-03 2013-06-04 Luciano Rabinowicz composition for preparation of energy drink and energy drink
US9464065B2 (en) 2011-03-24 2016-10-11 The Scripps Research Institute Compounds and methods for inducing chondrogenesis
CA2831143C (en) 2011-03-25 2019-05-21 Glaxosmithkline Intellectual Property Development Limited Cyclopropylamines as lsd1 inhibitors
WO2012147890A1 (en) 2011-04-27 2012-11-01 持田製薬株式会社 Novel azole derivative
EP2524918A1 (en) 2011-05-19 2012-11-21 Centro Nacional de Investigaciones Oncológicas (CNIO) Imidazopyrazines derivates as kinase inhibitors
EP2750671A2 (en) 2011-05-19 2014-07-09 Oryzon Genomics, S.A. Lysine demethylase inhibitors for thrombosis and cardiovascular diseases
WO2012156531A2 (en) 2011-05-19 2012-11-22 Oryzon Genomics, S.A. Lysine demethylase inhibitors for inflammatory diseases or conditions
WO2012168882A1 (en) 2011-06-07 2012-12-13 SPAI Group Ltd. Compositions and methods for improving stability and extending shelf life of sensitive food additives and food products thereof
AR086983A1 (en) 2011-06-20 2014-02-05 Incyte Corp DERIVATIVES OF AZETIDINIL FENIL, PIRIDIL OR PIRAZINIL CARBOXAMIDA AS JAK INHIBITORS
TW201311149A (en) 2011-06-24 2013-03-16 Ishihara Sangyo Kaisha Pesticide
EP2548877A1 (en) 2011-07-19 2013-01-23 MSD Oss B.V. 4-(5-Membered fused pyridinyl)benzamides as BTK-inhibitors
KR20140052018A (en) 2011-08-09 2014-05-02 다케다 야쿠힌 고교 가부시키가이샤 Cyclopropaneamine compound
SI2744330T1 (en) 2011-08-15 2020-11-30 University Of Utah Research Foundation Substituted (e)-n'-(1-phenylethylidene) benzohydrazide analogs as histone demethylase inhiitors
WO2013033688A1 (en) 2011-09-01 2013-03-07 The Brigham And Women's Hospital, Inc. Treatment of cancer
US9273343B2 (en) 2011-09-02 2016-03-01 Promega Corporation Compounds and methods for assaying redox state of metabolically active cells and methods for measuring NAD(P)/NAD(P)H
JP2015531401A (en) 2011-10-10 2015-11-02 ハー・ルンドベック・アクチエゼルスカベット PDE9I having imidazopyrazinone skeleton
RU2681211C2 (en) 2011-10-20 2019-03-05 Оризон Дженомикс С.А. (hetero)aryl cyclopropylamine compounds as lsd1 inhibitors
CL2014000988A1 (en) 2011-10-20 2014-11-03 Oryzon Genomics Sa Compounds derived from (aryl or heteroaryl) cyclopropylamide, lsd1 inhibitors; procedure to prepare them; pharmaceutical composition that includes them; and method to treat or prevent cancer, a neurological disease, a viral infection and viral reactivation after latency.
EP4074695A1 (en) 2011-10-20 2022-10-19 Oryzon Genomics, S.A. (hetero)aryl cyclopropylamine compounds as lsd1 inhibitors
ITMI20111971A1 (en) 2011-10-28 2013-04-29 Mesogenics Srl LSD-1 ENZYME INHIBITORS FOR THE INDUCTION OF OSTEOGENIC DIFFERENTIATION
EP2785183B1 (en) 2011-11-14 2018-12-19 Merck Sharp & Dohme Corp. Triazolopyridinone pde10 inhibitors
WO2013085877A1 (en) 2011-12-05 2013-06-13 Brandeis University Treatment of amyloidosis by compounds that regulate retromer stabilization
GB201205669D0 (en) 2012-03-30 2012-05-16 Agency Science Tech & Res Bicyclic heterocyclic derivatives as mnk2 and mnk2 modulators and uses thereof
CN102579381B (en) 2012-03-30 2013-07-10 河南中帅医药科技发展有限公司 Guanidine hydrochloride sustained release preparation and preparation method thereof
CN103373996A (en) 2012-04-20 2013-10-30 山东亨利医药科技有限责任公司 Bicyclic derivatives serving as CRTH2 receptor antagonist
WO2014002051A2 (en) 2012-06-28 2014-01-03 Novartis Ag Complement pathway modulators and uses thereof
CN102772444A (en) 2012-07-06 2012-11-14 周明千 Method for processing traditional Chinese medicinal ultramicro wall-broken oral tablet slices
GB201212513D0 (en) 2012-07-13 2012-08-29 Ucb Pharma Sa Therapeutic agents
CA2886187C (en) 2012-09-28 2020-04-14 Vanderbilt University Fused heterocyclic compounds as selective bmp inhibitors
CA2887203A1 (en) 2012-10-05 2014-04-10 Rigel Pharmaceuticals, Inc. 2,3-(hetero)aryl substituted pyridinyl compounds and their use as gdf-8 inhibitors
JP6325449B2 (en) 2012-10-12 2018-05-16 武田薬品工業株式会社 Cyclopropanamine compounds and uses thereof
US9757379B2 (en) 2012-11-14 2017-09-12 The Board Of Regents Of The University Of Texas System Inhibition of HIF-2α heterodimerization with HIF1β (ARNT)
JP6238908B2 (en) 2012-11-28 2017-11-29 京都府公立大学法人 LSD1 selective inhibitor having lysine structure
US9144555B2 (en) 2012-11-30 2015-09-29 Darlene E. McCord Hydroxytyrosol and oleuropein compositions for induction of DNA damage, cell death and LSD1 inhibition
EP2740474A1 (en) 2012-12-05 2014-06-11 Instituto Europeo di Oncologia S.r.l. Cyclopropylamine derivatives useful as inhibitors of histone demethylases kdm1a
MX360420B (en) 2012-12-19 2018-10-31 Wockhardt Ltd A stable aqueous composition comprising human insulin or an analogue or derivative thereof.
CN103054869A (en) 2013-01-18 2013-04-24 郑州大学 Application of amino dithio formic ester compound with triazolyl in preparing medicine taking LSD1 (Lysine Specificity Demethylase 1) as target
CN103933036B (en) 2013-01-23 2017-10-13 中国人民解放军军事医学科学院毒物药物研究所 2 Aryimidazoles simultaneously the acetamide derivative of [1,2 α] pyridine 3 prepare preventing and treating PTSD medicine in purposes
EP2956441A4 (en) 2013-02-18 2016-11-02 Scripps Research Inst Modulators of vasopressin receptors with therapeutic potential
US8558008B2 (en) 2013-02-28 2013-10-15 Dermira, Inc. Crystalline glycopyrrolate tosylate
WO2014164867A1 (en) 2013-03-11 2014-10-09 Imago Biosciences Kdm1a inhibitors for the treatment of disease
AU2014231768A1 (en) 2013-03-13 2015-09-24 Australian Nuclear Science And Technology Organisation Transgenic non-human organisms with non-functional TSPO genes
AU2014236348B2 (en) 2013-03-14 2018-05-10 Epizyme, Inc. Combination therapy for treating cancer
US20140343118A1 (en) 2013-03-14 2014-11-20 Duke University Methods of treatment using arylcyclopropylamine compounds
EP3003301B1 (en) 2013-05-30 2021-02-24 Board Of Regents Of The Nevada System Of Higher Education On Behalf Of The University Of Nevada, Las Vegas Novel suicidal lsd1 inhibitors targeting sox2-expressing cancer cells
SG11201510376QA (en) 2013-06-19 2016-01-28 Univ Utah Res Found Substituted (e)-n'-(1-phenylethylidene) benzohydrazide analogs as histone demethylase inhibitors
BR112015031864B1 (en) 2013-06-21 2022-02-15 Myokardia, Inc PYRIMIDINEDIONE COMPOUNDS AGAINST HEART CONDITIONS, PHARMACEUTICAL COMPOSITION AND USE
US9186391B2 (en) 2013-08-29 2015-11-17 Musc Foundation For Research Development Cyclic peptide inhibitors of lysine-specific demethylase 1
WO2015031564A2 (en) 2013-08-30 2015-03-05 University Of Utah Substituted-1h-benzo[d]imidazole series compounds as lysine-specfic demethylase 1 (lsd1) inhibitors
US9770514B2 (en) 2013-09-03 2017-09-26 ExxPharma Therapeutics LLC Tamper-resistant pharmaceutical dosage forms
CA2923990A1 (en) 2013-09-13 2015-03-19 Bayer Pharma Aktiengesellschaft Pharmaceutical compositions containing refametinib
KR101568724B1 (en) 2013-11-13 2015-11-12 서울대학교산학협력단 Novel compound, a preparing method thereof, and a use thereof as inhibitors of histone demethylase
EP4257591A3 (en) 2013-12-11 2023-11-22 Celgene Quanticel Research, Inc. Inhibitors of lysine specific demethylase-1
HUE045725T2 (en) 2014-02-13 2020-01-28 Incyte Corp Cyclopropylamines as lsd1 inhibitors
US9428470B2 (en) 2014-02-13 2016-08-30 Takeda Pharmaceutical Company Limited Heterocyclic compound
WO2015123437A1 (en) 2014-02-13 2015-08-20 Incyte Corporation Cyclopropylamines as lsd1 inhibitors
EP3392244A1 (en) 2014-02-13 2018-10-24 Incyte Corporation Cyclopropylamines as lsd1 inhibitors
US9346776B2 (en) 2014-02-13 2016-05-24 Takeda Pharmaceutical Company Limited Fused heterocyclic compound
LT3105218T (en) 2014-02-13 2019-12-10 Incyte Corp Cyclopropylamines as lsd1 inhibitors
CN103893163B (en) 2014-03-28 2016-02-03 中国药科大学 The application of 2-([1,1 '-biphenyl]-4-base) 2-oxoethyl 4-((the chloro-4-aminomethyl phenyl of 3-) is amino)-4-oxobutanoic acid esters in preparation LSD1 inhibitor medicaments
US9902722B2 (en) 2014-04-02 2018-02-27 Bristol-Myers Squibb Company Biaryl kinase inhibitors
CN106459024B (en) 2014-04-11 2019-11-05 武田药品工业株式会社 Cyclopropyl amine compounds and application thereof
EP2929884A1 (en) 2014-04-11 2015-10-14 Sanovel Ilac Sanayi ve Ticaret A.S. Pharmaceutical combinations of dabigatran and h2-receptor antagonists
EP2933002A1 (en) 2014-04-11 2015-10-21 Sanovel Ilac Sanayi ve Ticaret A.S. Pharmaceutical combinations of dabigatran and proton pump inhibitors
CN103961340B (en) 2014-04-30 2019-06-25 南通中国科学院海洋研究所海洋科学与技术研究发展中心 A kind of LSD1 inhibitor and its application
EP3148974B1 (en) 2014-05-30 2018-09-26 Istituto Europeo di Oncologia S.r.l. Cyclopropylamine compounds as histone demethylase inhibitors
CN104119280B (en) 2014-06-27 2016-03-16 郑州大学 Containing the pyrimidine derivatives of amino urea and Terminal Acetylenes structural unit, preparation method and application
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
WO2016007731A1 (en) 2014-07-10 2016-01-14 Incyte Corporation Imidazopyridines and imidazopyrazines as lsd1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
TW201613925A (en) 2014-07-10 2016-04-16 Incyte Corp Imidazopyrazines as LSD1 inhibitors
GB201417828D0 (en) 2014-10-08 2014-11-19 Cereno Scient Ab New methods and compositions
CN104173313B (en) 2014-08-25 2017-05-17 杭州朱养心药业有限公司 Rivaroxaban troche pharmaceutical composition
JP6653116B2 (en) 2014-08-27 2020-02-26 日本ケミファ株式会社 Olmesartan prodrug formulations
EA035185B1 (en) 2014-10-08 2020-05-12 Ф. Хоффманн-Ля Рош Аг Spirodiamine derivatives as aldosterone synthase inhibitors
CA2980395A1 (en) 2015-04-03 2016-10-06 Mutabilis Heterocyclic compounds and their use in preventing or treating bacterial infections
UY36601A (en) 2015-04-03 2016-09-30 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware INHIBITORS OF THE ENZYMATIC FUNCTION OF INDOLAMINE-2,3-DIOXYGENASE (IDO) AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
MA51438A (en) 2015-04-03 2021-04-14 Incyte Corp HETEROCYCLIC COMPOUNDS USED AS LSD1 INHIBITORS
MX2018001706A (en) 2015-08-12 2018-09-06 Incyte Corp Salts of an lsd1 inhibitor.
CN112656772B (en) 2015-10-15 2022-05-20 浙江东日药业有限公司 Rivaroxaban pharmaceutical composition
CA3009805C (en) 2015-12-29 2023-10-17 Mirati Therapeutics, Inc. Lsd1 inhibitors
JPWO2017130933A1 (en) 2016-01-25 2018-11-29 国立大学法人 熊本大学 Neurodegenerative disease therapeutic agent
CN109414410B (en) 2016-04-22 2022-08-12 因赛特公司 Formulations of LSD1inhibitors
BR112019014759A2 (en) 2017-01-18 2020-03-03 Vanderbilt University HETEROCYCLIC COMPOUNDS FOUND AS SELECTIVE BMP INHIBITORS
MX2019010354A (en) 2017-03-16 2019-10-22 Jiangsu Hengrui Medicine Co Heteroaryl[4,3-c]pyrimidine-5-amine derivative, preparation method therefor, and medical uses thereof.
AU2019297361A1 (en) 2018-07-05 2021-02-25 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
WO2020047198A1 (en) 2018-08-31 2020-03-05 Incyte Corporation Salts of an lsd1 inhibitor and processes for preparing the same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2013131609A1 (en) * 2012-03-07 2013-09-12 Merck Patent Gmbh Triazolopyrazine derivatives

Non-Patent Citations (71)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
ADAMO, A. ET AL.: "LSDI regulates the balance between self-renewal and differentiation in human embryonic stem cells", NAT CELL BIOL, vol. 13, no. 6, 2011, pages 652 - 9
ADAMO, A.; M.J. BARRERO; J.C. IZPISUA BELMONTE: "1$D1 and pluripotency: a new player in the network", CELL CYCLE, vol. 10, no. 19, 2011, pages 3215 - 6
ANAND, R.; R. MARMORSTEIN: "Structure and mechanism of lysine-specific demethylase enzymes", J BIOL CHEM, vol. 282, no. 49, 2007, pages 35425 - 9
BAUER, D.E.; S.C. KAMRAN; S.H. ORKIN: "Reawakeningfetal hemoglobin: prospects for new therapiesfor the beta-globin disorders", BLOOD, vol. 120, no. 15, 2012, pages 2945 - 53
BECK, B.; C. BLANPAIN: "Unravelling cancer stem cell potential", NAT REV CANCER, vol. 13, no. 10, 2013, pages 727 - 38
BENNANI-BAITI IDRISS M ET AL: "Lysine-specific demethylase 1 (LSD1/KDM1A/AOF2/BHC110) is expressed and is an epigenetic drug target in chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma", HUMAN PATHOLOGY, vol. 43, no. 8, 2 August 2012 (2012-08-02), pages 1300 - 1307, XP028929176, ISSN: 0046-8177, DOI: 10.1016/J.HUMPATH.2011.10.010 *
BENNANI-BAITI, I.M. ET AL.: "Lysine-specific demethylase I (LSDI/KDMIAlAOF2/BHC110) is expressed and is an epigenetic drug target in chondrosarcoma, Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma", HUM PATHOL, vol. 43, no. 8, 2012, pages 1300 - 7
BERGE ET AL., J. PHARM. SCI., vol. 66, no. 1, 1977, pages 1 - 19
CHEN, L; D.B. FLIES: "Molecular mechanisms of T cell co-stimulation and co-inhibition", NAT REV IMMUNOL, vol. 13, no. 4, 2013, pages 227 - 42
CHEN, Y. ET AL.: "Crystal structure of human histone lysine-specific demethylase I (LSD1)", PROC NATL ACAD SCI U S A, vol. 103, no. 38, 2006, pages 13956 - 61
CHEN, Y. ET AL.: "Lysine-specific histone demethylase I (LSDl): A potential molecular target for tumor therapy", CRIT REV EUKARYOT GENE EXPR, vol. 22, no. 1, 2012, pages 53 - 9
CHO, H.S. ET AL.: "Demethylation ofRB regulator MYPTI by histone demethylase LSDI promotes cell cycle progression in cancer cells", CANCER RES, vol. 71, no. 3, 2011, pages 655 - 60
CLEVERS, H.: "The cancer stem cell: premises, promises and challenges", NAT MED, vol. 17, no. 3, 2011, pages 313 - 9
CREA, F. ET AL.: "The emerging role of histone lysine demethylases in prostate cancer", MOL CANCER, no. 11, 2012, pages 52
DAWSON, M.A.; T. KOUZARIDES: "Cancer epigenetics: from mechanism to therapy", CELL, vol. 150, no. 1, 2012, pages 12 - 27
DING, J. ET AL.: "LSDI-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer", BR J CANCER, vol. 109, no. 4, 2013, pages 994 - 1003
FORNERIS, F. ET AL.: "Structural basis of LSD1-CoREST selectivity in histone H3 recognition", J BIOL CHEM, vol. 282, no. 28, 2007, pages 20070 - 4
GREAVES, P.; J.G. GRIBBEN: "The role of B7 family molecules in hematologic malignancy", BLOOD, vol. 121, no. 5, 2013, pages 734 - 44
HAKIMI, M.A. ET AL.: "A core-BRAF35 complex containing histone deacetylase mediates repression of neuronal-specific genes", PROC NATL ACAD SCI U S A, vol. 99, no. 11, 2002, pages 7420 - 5
HAN, H. ET AL.: "Synergistic re-activation of epigenetically silenced genes by combinatorial inhibition of DNMTs and LSDI in cancer cells", PLOS ONE, vol. 8, no. 9, 2013, pages E75136
HARRIS, W.J. ET AL.: "The histone demethylase KDMIA sustains the oncogenic potential ofMLL-AF9 leukemia stem cells", CANCER CELL, vol. 21, no. 4, 2012, pages 473 - 87
HAYAMI, S. ET AL.: "Overexpression ofLSD1 contributes to human carcinogenesis through chromatin regulation in various cancers", INT J CANCER, vol. 128, no. 3, 2011, pages 574 - 86
HUANG, J. ET AL.: "p53 is regulated by the lysine demethylase LSDl", NATURE, vol. 449, no. 7158, 2007, pages 105 - 8
K. BLOM: "Two-Pump At Column Dilution Configuration for Preparative LC-MS", J. COMBI. CHEM., vol. 4, 2002, pages 295
K. BLOM; B. GLASS; R. SPARKS; A. COMBS: "Preparative LCMS Purification: Improved Compound Specific Method Optimization", J. COMB. CHEM., vol. 6, 2004, pages 874 - 883
K. BLOM; B. GLASS; R. SPARKS; A. COMBS: "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", J. COMBI. CHEM., vol. 6, 2004, pages 874 - 883
K. BLOM; R. SPARKS; J. DOUGHTY; G. EVERLOF; T. HAQUE; A. COMBS: "Optimizing Preparative LC-MS Configurations and Methods for Parallel Synthesis Purification", J. COMBI. CHEM., vol. 5, 2003, pages 670
K. T. POTTS ET AL: "The mass spectra of somes-triazolo[4,3-a]pyrazines", ORGANIC MASS SPECTROMETRY, vol. 5, no. 6, 1 June 1971 (1971-06-01), pages 663 - 674, XP055210225, ISSN: 0030-493X, DOI: 10.1002/oms.1210050605 *
KAHL, P. ET AL.: "Androgen receptor coactivators lysine-specific histone demethylase land four and a half LIM domain protein 2 predict risk ofprostate cancer recurrence", CANCER RES, vol. 66, no. 23, 2006, pages 11341 - 7
KARL F. BLOM; BRIAN GLASS; RICHARD SPARKS; ANDREW P. COMBS: "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", J. COMBI. CHEM., vol. 6, no. 6, 2004, pages 874 - 883
KARYTINOS, A. ET AL.: "A novel mammalian flavin-dependent histone demethylase", J BIOL CHEM, vol. 284, no. 26, 2009, pages 17775 - 82
KONG, L. ET AL.: "Immunohistochemical expression ofRBP2 and LSD1 in papillary thyroid carcinoma", ROM J MORPHOL EMBRYOL, vol. 54, no. 3, 2013, pages 499 - 503
KONOVALOV, S.; I. GARCIA-BASSETS: "Analysis of the levels of lysine-specific demethylase I (LSD1) mRNA in human ovarian tumors and the effects of chemical LSDI inhibitors in ovarian cancer cell lines", J OVARIAN RES, vol. 6, no. 1, 2013, pages 75
KONTAKI, H.; I. TALIANIDIS: "Lysine methylation regulates E2Fl-induced cell death", MOL CELL, vol. 39, no. 1, 2010, pages 152 - 60
KOOISTRA, S.M.; K. HELIN: "Molecular mechanisms and potential functions of histone demethylases", NAT REV MOL CELL BIOL, vol. 13, no. 5, 2012, pages 297 - 311
KUROYANAGI J I ET AL: "1,3-Benzoxazole-4-carbonitrile as a novel antifungal scaffold of beta-1,6-glucan synthesis inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 18, no. 21, 1 November 2010 (2010-11-01), pages 7593 - 7606, XP027415413, ISSN: 0968-0896, [retrieved on 20100925] *
KUROYANAGI J I ET AL: "Novel antifungal agents: Triazolopyridines as inhibitors of beta-1,6-glucan synthesis", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 18, no. 16, 15 August 2010 (2010-08-15), pages 5845 - 5854, XP027192826, ISSN: 0968-0896, [retrieved on 20100701] *
LEE, M.G. ET AL.: "Functional interplay between histone demethylase and deacetylase enzymes", MOL CELL BIOL, vol. 26, no. 17, 2006, pages 6395 - 402
LIANG, Y. ET AL.: "Targeting the JMJD2 histone demethylases to epigenetically control herpesvirus infection and reactivation from latency. Sci Transl Med, 2013. 5(167): p. 167ra5; Liang, Y., et al., Inhibition of the histone demethylase LSD1 blocks alpha-herpesvirus lytic replication and reactivation from latency", NAT MED, vol. 15, no. 11, 2009, pages 1312 - 7
LIM, S. ET AL.: "Lysine-specific demethylase I (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology", CARCINOGENESIS, vol. 31, no. 3, 2010, pages 512 - 20
LUND, A.H.; M. VAN LOHUIZEN: "Epigenetics and cancer", GENES DEV, vol. 18, no. 19, 2004, pages 2315 - 35
LV, T. ET AL.: "Over-expression of LSDI promotes proliferation, migration and invasion in non-small cell lung cancer", PLOS ONE, vol. 7, no. 4, 2012, pages E35065
LYNCH, J.T. ET AL.: "CD86 expression as a surrogate cellular biomarker for pharmacological inhibition of the histone demethylase lysine-specific demethylase 1", ANAL BIOCHEM, vol. 442, no. 1, 2013, pages 104 - 6
METZGER, E. ET AL.: "LSDI demethylates repressive histone marks to promote androgen-receptor-dependent transcription", NATURE, vol. 437, no. 7057, 2005, pages 436 - 9
MOSAMMAPARAST, N.; Y. SHI: "Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases", ANNU REV BIOCHEM, vol. 79, 2010, pages 155 - 79
NAGASAWA, J. AM. CHEM. SOC., vol. 131, no. 42, 2009, pages 15080
P. G. M. WUTS; T. W. GREENE: "Protective Groups in Organic Synthesis", 2006, WILEY & SONS, INC.
PORTELA, A.; M. ESTELLER: "Epigenetic modifications and human disease", NAT BIOTECHNOL, vol. 28, no. 10, 2010, pages 1057 - 68
SAKANE, N. ET AL.: "Activation of HIV transcription by the viral Tat protein requires a demethylation step mediated by lysine-specific demethylase I (LSDI/KDMI )", PLOS PATHOG, vol. 7, no. 8, 2011, pages E1002184
SANKARAN, V.G.; S.H. ORKIN: "The switch from fetal to adult hemoglobin", COLD SPRING HARB PERSPECT MED, vol. 3, no. 1, 2013, pages AOL 1643
SAREDDY, G.R. ET AL.: "KDMI is a novel therapeutic targetfor the treatment ofgliomas", ONCOTARGET, vol. 4, no. 1, 2013, pages 18 - 28
SCHENK, T. ET AL.: "Inhibition of the LSDI (KDMIA) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia", NAT MED, vol. 18, no. 4, 2012, pages 605 - 11
SCHULTE, J.H. ET AL.: "Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma: implications for therapy", CANCER RES, vol. 69, no. 5, 2009, pages 2065 - 71
SERCE, N. ET AL.: "Elevated expression of LSD1 (Lysine-specific demethylase 1) during tumour progression from pre-invasive to invasive ductal carcinoma of the breast", BMC CLIN PATHOL, vol. 12, 2012, pages 13
SHEN, H; P.W. LAIRD: "Interplay between the cancer genome and epigenome", CELL, vol. 153, no. 1, 2013, pages 38 - 55
SHI, L. ET AL.: "Lysine-specific demethylase 1 is a therapeutic target for fetal hemoglobin induction", NAT MED, vol. 19, no. 3, 2013, pages 291 - 4
SHI, Y. ET AL.: "Histone demethylation mediated by the nuclear amine oxidase homolog LSDl", CELL, vol. 119, no. 7, 2004, pages 941 - 53
SINGH, M.M. ET AL.: "Inhibition of LSDI sensitizes glioblastoma cells to histone deacetylase inhibitors", NEURO ONCOL, vol. 13, no. 8, 2011, pages 894 - 903
STAHL ET AL.: "Handbook ø/ Pharmaceutica/ Salts: Properties, Selection, and Use", 2002, WILEY
STAVROPOULOS, P.; G. BLOBEL; A. HOELZ: "Crystal structure and mechanism of human lysine-specific demethylase-l", NAT STRUCT MOL BIOL, vol. 13, no. 7, 2006, pages 626 - 32
SUIKKI, H.E. ET AL.: "Genetic alterations and changes in expression of histone demethylases in prostate cancer", PROSTATE, vol. 70, no. 8, 2010, pages 889 - 98
SUN, G. ET AL.: "Histone demethylase LSDI regulates neural stem cell proliferation", MOL CELL BIOL, vol. 30, no. 8, 2010, pages 1997 - 2005
WALDMANN, T.; R. SCHNEIDER: "Targeting histone modyications--epigenetics in cancer", CURR OPIN CELL BIOL, vol. 25, no. 2, 2013, pages 184 - 9
WANG, J. ET AL.: "Novel histone demethylase L$D1 inhibitors selectively target cancer cells with pluripotent stem cell properties", CANCER RES, vol. 71, no. 23, 2011, pages 7238 - 49
WANG, J. ET AL.: "The lysine demethylase LSDI (KDMI) is required for maintenance of global DNA methylation", NAT GENET, vol. 41, no. 1, 2009, pages 125 - 9
XU, J. ET AL.: "Corepressor-dependent silencing offetal hemoglobin expression by BCL11A", PROC NATL ACAD SCI U S A, vol. 110, no. 16, 2013, pages 6518 - 23
YANG, J. ET AL.: "Reversible methylation ofpromoter-bound STAT3 by histone-modifying enzymes", PROC NATL ACAD SCI U S A, vol. 107, no. 50, 2010, pages 21499 - 504
YOU, A. ET AL.: "CoREST is an integral component of the C REST- human histone deacetylase complex", PROC NATL ACAD SCI U S A, vol. 98, no. 4, 2001, pages 1454 - 8
YU, Y. ET AL.: "High expression of lysine-specific demethylase I correlates with poor prognosis of patients with esophageal squamous cell carcinoma", BIOCHEM BIOPHYS RES COMMUN, vol. 437, no. 2, 2013, pages 192 - 8
ZHANG, X. ET AL.: "Pluripotent Stem Cell Protein Sox2 Confers Sensitivity to L$D1 Inhibition in Cancer Cells", CELL REP, vol. 5, no. 2, 2013, pages 445 - 57

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9994546B2 (en) 2014-02-13 2018-06-12 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11247992B2 (en) 2014-02-13 2022-02-15 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US11155532B2 (en) 2014-02-13 2021-10-26 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10717737B2 (en) 2014-02-13 2020-07-21 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US9670210B2 (en) 2014-02-13 2017-06-06 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10676457B2 (en) 2014-02-13 2020-06-09 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10513493B2 (en) 2014-02-13 2019-12-24 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10174030B2 (en) 2014-02-13 2019-01-08 Incyte Corporation Cyclopropylamines as LSD1 inhibitors
US10640503B2 (en) 2014-07-10 2020-05-05 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US9695167B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10968221B2 (en) 2014-07-10 2021-04-06 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US9695180B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10556908B2 (en) 2014-07-10 2020-02-11 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US9758523B2 (en) 2014-07-10 2017-09-12 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US9695168B2 (en) 2014-07-10 2017-07-04 Incyte Corporation Substituted imidazo[1,5-α]pyridines and imidazo[1,5-α]pyrazines as LSD1 inhibitors
US10047086B2 (en) 2014-07-10 2018-08-14 Incyte Corporation Imidazopyridines and imidazopyrazines as LSD1 inhibitors
US10112950B2 (en) 2014-07-10 2018-10-30 Incyte Corporation Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10125133B2 (en) 2014-07-10 2018-11-13 Incyte Corporation Substituted [1,2,4]triazolo[1,5-a]pyridines and substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10138249B2 (en) 2014-07-10 2018-11-27 Incyte Corporation Triazolopyridines and triazolopyrazines as LSD1 inhibitors
AU2016243939B2 (en) * 2015-04-03 2020-09-03 Incyte Holdings Corporation Heterocyclic compounds as LSD1 inhibitors
US10800779B2 (en) 2015-04-03 2020-10-13 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
WO2016161282A1 (en) * 2015-04-03 2016-10-06 Incyte Corporation Heterocyclic compounds as lsd1 inhibitors
EP3626720A1 (en) * 2015-04-03 2020-03-25 Incyte Corporation Heterocyclic compounds as lsd1 inhibitors
US11401272B2 (en) 2015-04-03 2022-08-02 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
US9944647B2 (en) 2015-04-03 2018-04-17 Incyte Corporation Heterocyclic compounds as LSD1 inhibitors
WO2016198649A1 (en) 2015-06-12 2016-12-15 Oryzon Genomics, S.A. Biomarkers associated with lsd1 inhibitors and uses thereof
WO2017013061A1 (en) 2015-07-17 2017-01-26 Oryzon Genomics, S.A. Biomarkers associated with lsd1 inhibitors and uses thereof
US10723700B2 (en) 2015-08-12 2020-07-28 Incyte Corporation Salts of an LSD1 inhibitor
US11498900B2 (en) 2015-08-12 2022-11-15 Incyte Corporation Salts of an LSD1 inhibitor
US10329255B2 (en) 2015-08-12 2019-06-25 Incyte Corporation Salts of an LSD1 inhibitor
JP2019508447A (en) * 2016-03-15 2019-03-28 セルジーン クオンティセル リサーチ,インク. Histone demethylase inhibitors
US10265279B2 (en) 2016-03-15 2019-04-23 Oryzon Genomics, S.A. Combinations of LSD1 inhibitors for use in the treatment of solid tumors
WO2017157825A1 (en) 2016-03-15 2017-09-21 F. Hoffmann-La Roche Ag Combinations of lsd1 inhibitors for use in the treatment of solid tumors
WO2017158136A1 (en) 2016-03-16 2017-09-21 Oryzon Genomics, S.A. Methods to determine kdm1a target engagement and chemoprobes useful therefor
US10166221B2 (en) 2016-04-22 2019-01-01 Incyte Corporation Formulations of an LSD1 inhibitor
CN106432248A (en) * 2016-09-27 2017-02-22 郑州大学 Pyrimidine and triazole containing LSD1 inhibitor and preparation method and application thereof
WO2018083189A1 (en) 2016-11-03 2018-05-11 Oryzon Genomics, S.A. Biomarkers for determining responsiveness to lsd1 inhibitors
CN106928235A (en) * 2017-05-03 2017-07-07 郑州大学 The LSD1 of triazole containing pyrimido inhibitor, its preparation method and application
WO2019025588A1 (en) 2017-08-03 2019-02-07 Oryzon Genomics, S.A. Methods of treating behavior alterations
KR20190030192A (en) 2017-09-13 2019-03-21 한미약품 주식회사 Pyrazole derivative compound and use thereof
US11254676B2 (en) 2017-09-13 2022-02-22 Hanmi Pharm. Co., Ltd. Pyrazole derivative compound and use thereof
WO2019054766A1 (en) 2017-09-13 2019-03-21 Hanmi Pharm. Co., Ltd. Pyrazole derivative compound and use thereof
WO2019068326A1 (en) 2017-10-05 2019-04-11 Université D'aix-Marseille Lsd1 inhibitors for the treatment and prevention of cardiomyopathies
KR20190040763A (en) 2017-10-11 2019-04-19 한미약품 주식회사 Pyrazolopyridine derivative compound and use thereof
KR20190040783A (en) 2017-10-11 2019-04-19 한미약품 주식회사 Pyrazole derivatives as Lysine-specific histone demethylase-1 inhibitors
US11673894B2 (en) 2018-02-27 2023-06-13 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
US11168089B2 (en) 2018-05-18 2021-11-09 Incyte Corporation Fused pyrimidine derivatives as A2A / A2B inhibitors
US11873304B2 (en) 2018-05-18 2024-01-16 Incyte Corporation Fused pyrimidine derivatives as A2A/A2B inhibitors
US11161850B2 (en) 2018-07-05 2021-11-02 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
US10968200B2 (en) 2018-08-31 2021-04-06 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11512064B2 (en) 2018-08-31 2022-11-29 Incyte Corporation Salts of an LSD1 inhibitor and processes for preparing the same
US11390624B2 (en) 2019-01-29 2022-07-19 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
US11884665B2 (en) 2019-01-29 2024-01-30 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
WO2020159285A1 (en) 2019-02-01 2020-08-06 한미약품 주식회사 Imidazopyridine derivative compounds and use of same
KR20200096157A (en) 2019-02-01 2020-08-11 한미약품 주식회사 Imidazopyridine derivative compound and use thereof
WO2020188089A1 (en) 2019-03-20 2020-09-24 Oryzon Genomics, S.A. Methods of treating attention deficit hyperactivity disorder using kdm1a inhibitors such as the compound vafidemstat
WO2020188090A1 (en) 2019-03-20 2020-09-24 Oryzon Genomics, S.A. Methods of treating borderline personality disorder
WO2021004610A1 (en) 2019-07-05 2021-01-14 Oryzon Genomics, S.A. Biomarkers and methods for personalized treatment of small cell lung cancer using kdm1a inhibitors
WO2022053520A1 (en) 2020-09-08 2022-03-17 Université D'aix Marseille Lsd1 inhibitors for use in the treatment and prevention of fibrosis of tissues
EP3964204A1 (en) 2020-09-08 2022-03-09 Université d'Aix-Marseille Lsd1 inhibitors for use in the treatment and prevention of fibrosis of tissues
WO2022214303A1 (en) 2021-04-08 2022-10-13 Oryzon Genomics, S.A. Combinations of lsd1 inhibitors for treating myeloid cancers
CN115772170A (en) * 2021-12-03 2023-03-10 徐诺药业(南京)有限公司 Pyrazolo [1,5-a ] pyridine derivative and preparation method and application thereof
WO2023217784A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating nf1-mutant tumors using lsd1 inhibitors
WO2023217758A1 (en) 2022-05-09 2023-11-16 Oryzon Genomics, S.A. Methods of treating malignant peripheral nerve sheath tumor (mpnst) using lsd1 inhibitors

Also Published As

Publication number Publication date
US9695167B2 (en) 2017-07-04
US20170369488A1 (en) 2017-12-28
US20200024277A1 (en) 2020-01-23
US20160009711A1 (en) 2016-01-14
US10125133B2 (en) 2018-11-13
US20190119272A1 (en) 2019-04-25
US10968221B2 (en) 2021-04-06

Similar Documents

Publication Publication Date Title
US10968221B2 (en) Substituted [1,2,4]triazolo[1,5-a]pyrazines as LSD1 inhibitors
US10138249B2 (en) Triazolopyridines and triazolopyrazines as LSD1 inhibitors
US10513493B2 (en) Cyclopropylamines as LSD1 inhibitors
US10676457B2 (en) Cyclopropylamines as LSD1 inhibitors
US10556908B2 (en) Substituted imidazo[1,2-a]pyrazines as LSD1 inhibitors
US10300051B2 (en) Cyclopropylamines as LSD1 inhibitors
AU2016243939B2 (en) Heterocyclic compounds as LSD1 inhibitors
KR102430144B1 (en) 1H-PYRROLO[2,3-c]PYRIDIN-7(6H)-ONES AND PYRAZOLO[3,4-c]PYRIDIN-7(6H)-ONES AS INHIBITORS OF BET PROTEINS

Legal Events

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

Ref document number: 15739467

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15739467

Country of ref document: EP

Kind code of ref document: A1