Methods of using indazole-3-carb oxamides and their use as Wnt/B-catenin signaling pathway inhibitors

Information

  • Patent Grant
  • 10806726
  • Patent Number
    10,806,726
  • Date Filed
    Monday, October 23, 2017
    6 years ago
  • Date Issued
    Tuesday, October 20, 2020
    3 years ago
Abstract
This disclosure features the use of one or more indazole-3-carboxamide compounds or salts or analogs thereof, in the treatment of one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease; and/or for promoting wound healing. The methods include administering to a subject (e.g., a subject in need thereof) a therapeutically effective amount of one or more indazole-3-carboxamide compounds or salts or analogs thereof as described anywhere herein.
Description
BACKGROUND
Technical Field

This disclosure features the use of one or more indazole-3-carboxamide compounds or salts or analogs thereof, in the treatment of one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease; and/or for promoting wound healing. The methods include administering to a subject (e.g., a subject in need thereof) a therapeutically effective amount of one or more indazole-3-carboxamide compounds or salts or analogs thereof as described anywhere herein.


Background

Pattern formation is the activity by which embryonic cells form ordered spatial arrangements of differentiated tissues. Speculation on the mechanisms underlying these patterning effects usually centers on the secretion of a signaling molecule that elicits an appropriate response from the tissues being patterned. More recent work aimed at the identification of such signaling molecules implicates secreted proteins encoded by individual members of a small number of gene families.


The Wnt growth factor family includes more than 10 genes identified in the mouse and at least 19 genes identified in the human. Members of the Wnt family of signaling molecules mediate many important short- and long-range patterning processes during invertebrate and vertebrate development. The Wnt signaling pathway is known for its important role in the inductive interactions that regulate growth and differentiation, and plays important roles in the homeostatic maintenance of post-embryonic tissue integrity. Wnt stabilizes cytoplasmic β-catenin, which stimulates the expression of genes including c-myc, c jun, fra-1, and cyclin Dl. In addition, misregulation of Wnt signaling can cause developmental defects and is implicated in the genesis of several human cancers. More recently, the Wnt pathway has been implicated in the maintenance of stem or progenitor cells in a growing list of adult tissues that now includes skin, blood, gut, prostate, muscle and the nervous system.


Pathological activation of the Wnt pathway is also believed to be the initial event leading to colorectal cancer in over 85% of all sporadic cases in the Western world. Activation of the Wnt pathway has also been extensively reported for hepatocellular carcinoma, breast cancer, ovarian cancer, pancreatic cancer, melanomas, mesotheliomas, lymphomas and leukemias. In addition to cancer, inhibitors of the Wnt pathway can be used for stem cell research or for the treatment of any diseases characterized by aberrant Wnt activation such as diabetic retinopathy, pulmonary fibrosis, rheumatoid arthritis, scleroderma as well as mycotic and viral infections and bone and cartilage diseases. As such, it is a therapeutic target that is of great interest to the field.


In addition to cancer, there are many cases of genetic diseases due to mutations in Wnt signaling components. Examples of some of the many diseases are Alzheimer's disease [Proc. Natl. Acad. Sci. USA (2007), 104(22), 9434-9], osteoarthritis, polyposis coli [Science (1991), 253(5020), 665-669], bone density and vascular defects in the eye (osteoporosis-pseudoglioma syndrome, OPPG) [N. Engl. J. Med. (2002), 346(20), 1513-21], familial exudative vitreoretinopathy [Hum. Mutat. (2005), 26(2), 104-12], retinal angiogenesis [Nat. Genet. (2002), 32(2), 326-30], early coronary disease [Science (2007), 315(5816), 1278-82], tetra-amelia syndrome [Am. J Hum. Genet. (2004), 74(3), 558-63], Müllerian-duct regression and virilization [Engl. J. Med. (2004), 351(8), 792-8], SERKAL syndrome [Am. J Hum. Genet. (2008), 82(1), 39-47], diabetes mellitus type 2 [Am. J. Hum. Genet. (2004), 75(5), 832-43; N. Engl. J. Med. (2006), 355(3), 241-50], Fuhrmann syndrome [Am. J Hum. Genet. (2006), 79(2), 402-8], Al-Awadi/Raas-Rothschild/Schinzel phocomelia syndrome [Am. J Hum. Genet. (2006), 79(2), 402-8], odonto-onycho-dermal dysplasia [Am. J Hum. Genet. (2007), 81(4), 821-8], obesity [Diabetologia (2006), 49(4), 678-84], split-hand/foot malformation [Hum. Mol. Genet. (2008), 17(17), 2644-53], caudal duplication syndrome [Am. J Hum. Genet. (2006), 79(1), 155-62], tooth agenesis [Am. J Hum. Genet. (2004), 74(5), 1043-50], Wilms tumor [Science (2007), 315(5812), 642-5], skeletal dysplasia [Nat. Genet. (2009), 41(1), 95-100], focal dermal hypoplasia [Nat. Genet. (2007), 39(7), 836-8], autosomal recessive anonychia [Nat. Genet. (2006), 38(11), 1245-7], neural tube defects [N. Engl. J Med. (2007), 356(14), 1432-7], alpha-thalassemia (ATRX) syndrome [The Journal of Neuroscience (2008), 28(47), 12570-12580], fragile X syndrome [PLoS Genetics (2010), 6(4), e1000898], ICF syndrome, Angelman syndrome [Brain Research Bulletin (2002), 57(1), 109-119], Prader-Willi syndrome [Journal of Neuroscience (2006), 26(20), 5383-5392], Beckwith-Wiedemann Syndrome [Pediatric and Developmental Pathology (2003), 6(4), 299-306] and Rett syndrome.


Regulation of cell signaling by the Wnt signaling pathway is critical for the formation of neuronal circuits. Wnt pathway modulates in neural tissue, among other things, axon pathfinding, dendritic development, and synaptic assembly. Through different receptors, Wnt pathway activates and/or regulates diverse signaling pathways and other processes that lead to local changes on the cytoskeleton or global cellular changes involving nuclear function. Recently, a link between neuronal activity, essential for the formation and refinement of neuronal connections, and Wnt signaling has been uncovered. Indeed, neuronal activity regulates the release of various Wnt proteins and the localization of their receptors. Wnt pathway mediates synaptic structural changes induced by neuronal activity or experience. Evidence suggests that dysfunction in Wnt signaling contributes to neurological disorders [Brain Research Reviews (2000), 33(1), 1-12; Oncogene (2006) 25(57), 7545-7553; Molecular Neurodegeneration (2008), 3, 9; Neurobiology of Disease (2010), 38(2), 148-153; Journal of Neurodevelopmental Disorders (2011), 3(2), 162-174 and Cold Spring Harbor Perspectives in Biology February (2012), 4(2)].


Tendinopathies are chronic disorders or injuries of the tendons, that typically result from gradual wear and tear to the tendon, e.g., from overuse or aging, and leading to is tendon degeneration, weakness, tearing, and pain. Individuals who tend to make multiple, repeated motions in their jobs, sports, or regular daily activities tend to be more likely to develop tendinopathies. Tendinopathy usually causes pain, stiffness, and loss of strength in the affected area.


Skin disorders are common afflictions for many people. Some of the most common are dermatitis (also known as eczema) and psoriasis. Both dermatitis and psoriasis can cause serious physical and/or psychological suffering to the subject regardless of the location on the body that these conditions occur.


WO 2013/040215 describes indazole-3-carboxamides having Formula (I) and their use as Wnt/B-catenin signaling pathway inhibitors.




embedded image


SUMMARY

This disclosure features the use of one or more indazole-3-carboxamide compounds or salts or analogs thereof, in the treatment of one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease; and/or for promoting wound healing. The methods include administering to a subject (e.g., a subject in need thereof) a therapeutically effective amount of one or more indazole-3-carboxamide compounds or salts or analogs thereof as described anywhere herein.


One embodiment disclosed herein includes administering an indazole-3-carboxamide compound having the structure of Formula I:




embedded image



as well as prodrugs and pharmaceutically acceptable salts thereof.


In some embodiments of Formula (I):


R1, R2 and R4 are independently selected from the group consisting of H, C1-9 alkyl, halide, —N(R10)2, —XR10, CN, —OCF3 and —CF3;


R3 is selected from the group consisting of carbocyclylR6, heterocyclylR6, arylR6 and heteroarylR6;


R5 is selected from the group consisting of —(C1-9 alkyl)ncarbocyclylR7, —(C1-9 alkyl)nheterocyclylR7, —(C1-9 alkyl)narylR7 and —(C1-9 alkyl)nheteroarylR7; each R6 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR10, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8, —(C1-9 alkyl)nheteroarylR8, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9 alkyl)nN(R10)2, —(C1-9 alkyl)nN(R10)SO2R11 and —SO2R11;


each R7 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR1′, —(C1-9 alkyl)ncarbocyclylR9, —(C1-9 alkyl)nheterocyclylR9, —(C1-9 alkyl)narylR9, —(C1-9 alkyl)nheteroarylR9, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9 alkyl)nN(R10)2, —(C1-9 alkyl)nN(R10)SO2R11 and —SO2R11;


each R8 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9 alkyl)nN(R12)2, —(C1-9 alkyl)nN(R12)SO2R13 and —SO2R13;


each R9 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9 alkyl)nN(R12)2, —(C1-9 alkyl)nN(R12)SO2R13 and —SO2R13;


each R10 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8;


each R11 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8;


each R12 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9 alkyl)naryl and —(C1-9 alkylNheteroaryl;


each R13 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9 alkylNaryl and —(C1-9alkyl)nheteroaryl; each R14 is independently selected from the group consisting of H, C1-3 alkyl, carbocyclyl and aryl;


each X is selected from the group consisting of a bond, —O— and —S—; and each n is 0 or 1.


In another embodiment of Formula (I):


R1, R2 and R4 are independently selected from the group consisting of H, C1-9 alkyl, halide, —N(R10)2, —XR10, CN, —OCF3 and —CF3;


R3 is selected from the group consisting of carbocyclylR6, heterocyclylR6, arylR6 and heteroarylR6;


in some embodiments, it is provided that when R3 is heteroaryl, the heteroaryl is not selected from the group consisting of isoquinoline, 1H-pyrrolo[2,3-c]pyridine and tetrazole;


R5 is selected from the group consisting of —(C1-9 alkyl)ncarbocyclylR7, —(C1-9 alkyl)nheterocyclylR7, —(C1-9 alkyl)narylR7 and —(C1-9 alkyl)nheteroaryl7;


in some embodiments, it is provided that R5 is not 4-pyridylR7 when R1, R2 and R4 are H, R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6, 2-pyridylR6, phenylR6, thiazoleR6, imidazoleR6, pyrimidineR6, oxazoleR6,




embedded image



and R6 and R7 are both H.


in some embodiments, it is provided that R5 is not —(CH2)(3-pyridyl)R7 when R1, R2 and R4 are H, R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6 and thiazoleR6, and R6 and R7 are both H;


in some embodiments, it is provided that R5 is not phenylR7 when R1, R2 and R4 are H, R3 is 4-pyridylR6 and R6 and R7 are both H;


in some embodiments, it is provided that R3 is not 3-pyridylR6 when R1, R2 and R4 are H, R5 is selected from the group consisting of phenylR7,




embedded image



and R6 and R7 are both H;


in some embodiments, it is provided that R3 is not oxazoleR6 when R1, R2 and R4 are H, R5 is selected from the group consisting of




embedded image



and R6 is H;


in some embodiments, it is provided that R3 is not thiazoleR6 when R1, R2 and R4 are H, R5 is selected from the group consisting of




embedded image



and R is H;


each R6 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR10, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8, —(C1-9 alkyl)nheteroarylR8, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9 alkyl)nN(R10)2, —(C1-9 alkyl)nN(R10)SO2R11 and —SO2R11;


each R7 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR10, —(C1-9 alkyl)ncarbocyclylR9, —(C1-9 alkyl)nheterocyclylR9, —(C1-9 alkyl)narylR9, —(C1-9 alkyl)nheteroarylR9, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9 alkyl)nN(R10)2, —(C1-9 alkyl)nN(R10)SO2R11 and —SO2R11;


each R8 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9 alkyl)nN(R12)2, —(C1-9 alkyl)nN(R12)SO2R13 and —SO2R13;


each R9 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9 alkyl)nN(R12)2, —(C1-9 alkyl)nN(R12)SO2R13 and —SO2R13;


each R10 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8;


each R11 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8;


each R12 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9 alkyl)naryl and —(C1-9 alkyl)nheteroaryl;


each R13 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9alkyl)naryl and —(C1-9alkyl)nheteroaryl;


each R14 is independently selected from the group consisting of H, C1-3 alkyl, carbocyclyl and aryl;


each X is selected from the group consisting of a bond, —O— and —S—; and


each n is 0 or 1.


Some embodiments include administering stereoisomers and pharmaceutically acceptable salts of a compound of general Formula (I).


Some embodiments include administering pro-drugs of a compound of general Formula (I).


Some embodiments include administering pharmaceutical compositions comprising a compound of general Formula (I) or in a pharmaceutically acceptable carrier, diluent, or excipient.


It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 depicts a plot showing that exposure of a human colorectal cancer cell line (SW480) to compound 175 inhibited Wnt/3-catenin activity in these cells in a dose-dependent manner (EC50=152.9 nM). The human colorectal cancer cell line (SW480) with constitutive activation of the Wnt pathway and engineered to express a Wnt responsive promoter linked to luciferase were treated with compound 175 attest concentrations of 10, 3, 1, 0.3, 0.1, 0.03, 0.01, 0.003, 0.001 and 0.0003 μM.



FIGS. 2A-C depict plots of the percentage of human mesenchymal stem cells (hMSC) expressing SCXA, tenacinC, and tenomodulin, respectively vs. concentration of compound 175. Human Mesenchymal Stem Cells were treated with compound 175 at test concentrations of 750, 333.3, 166.6, 83.3, 41.7, 21.7, 10.8 and 5.8 nM. Exposure of the cells to compound 175 for 7 days induced the expression of markers of tenocyte differentiation—SCXA, TenacinC and Tenomodulin in a dose-dependent manner (EC50=139-189 nM).



FIG. 3 are images of the expression of SCXA, tenacinC, and tenomodulin in hMSCs treated with DMSO, bone morphogenic protein (BMP) and fibroblast growth factor (FGF), and compound 175 at 333 nM concentration.



FIGS. 4A-B depict plots of tumor necrosis factor alpha concentration in THP-1 cells vs. the logarithm of the concentration of compound 175 the THP-1 cells were exposed to, and interleukin-6 concentration in THP-1 cells vs. the logarithm of the concentration of compound 175 the THP-1 cells were exposed to, respectively. THP-1 cells, a human monocyte cell line (6×10e4 cells/well in a 96-well plate) were treated with DMSO (vehicle) or compound 175 at concentrations of 10, 3.5, 1, 0.5, 0.1, 0.035, 0.01, 0.005 μM. After 2 hours, treated cells were stimulated with EPS (50 ng/mL for 5 hours [TNFα] and 500 ng/ml for 22 hours [IL6]) at 37° C. to induce production of the cytokines. Supernatants (diluted 1:1 or 1:4) were collected and analyzed for TNFα and IL6 levels using the TNFα and IL6 ELISA kits. Inhibition profile and EC50 was calculated using Prism 5. Representative images indicate that exposure of the cells to compound 175 inhibited the production of TNFα (4A) and IL6 (4B) in THP-1 cells in a dose-dependent manner, with an average EC50 of 342-547 nM for TNFα and 356-629 nM for IL6 (2 independent assays and 3 replicates per assay).



FIG. 5 is a grid of images of a rat Achilles tendon; a rat Achilles tendon treated with a collagenase with a vehicle; a rat Achilles tendon treated with a collagenase, compound 175, and benzyl alcohol; and a rat Achilles tendon treated with a collagenase and compound 175. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) or sham needle puncture for control in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle control or compound 175 (10 mg/ml). 24 hours after the last dose, tendons were isolated, fixed with 10% buffered formalin, sectioned and stained with H&E. Group 1 (sham injection), Group 2 (Collagenase-Vehicle), Group 3 (Collagenase-compound 175 with 1% BA) and Group 4 (Collagenase-compound 175 with 0.5% Tween 80, without BA).



FIG. 6 is a plot of tendon histology scores of rat Achilles tendon; a rat Achilles tendon treated with a collagenase with a vehicle; a rat Achilles tendon treated with a collagenase, compound 175, and benzyl alcohol; and a rat Achilles tendon treated with a collagenase and compound 175. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) or sham needle puncture for control in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle control or compound 175 (10 mg/ml). 24 hours after the last dose, tendons were isolated, fixed with 10% buffered formalin, sectioned and stained with H&E. Group 1 (sham injection), Group 2 (Collagenase-Vehicle), Group 3 (Collagenase-compound 175 with 1% BA) and Group 4 (Collagenase-compound 175 with 0.5% Tween 80, without BA). Blinded histology scoring of tendon injury and inflammation indicated that compound 175 with or without BA preservative significantly ameliorated collagenase-induced tendinopathy (**p<0.01 and *p<0.05 respectively) by student's t test. For this study, a total of 64 sections were scored for Group 1 and ˜96 sections each for Group 2-4.



FIG. 7 is series of plots of plasma concentrations of KC/GRO in rats vs. the number of days elapsed after administration of a vehicle, compound 175 with benzyl alcohol, and compound 175. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle control or compound 175 (10 mg/ml). Blood was collected at various timepoints and plasma was analyzed for KC/GRO by ELISA. Measurement of inflammatory cytokine KC/GRO in the plasma of rats from Group 2 (Collagenase-Vehicle), Group 3 (Collagenase-compound 175 10 mg/mL with 1% BA) and Group 4 (Collagenase-compound 175 10 mg/mL without BA, with 0.5% Tween 80). Compound 175 with or without BA preservative significantly decreased the levels of KC/GRO in plasma (p<0.05, student's t test as indicated in Table 3), indicating an anti-inflammatory effect of compound 175. These data were generated from 3 replicates per timepoint per group (n=3).



FIG. 8 is a grid of images of a rat Achilles tendon; a rat Achilles tendon treated with a collagenase with a vehicle; a rat Achilles tendon treated with a collagenase, 3 mg/ml compound 175, and benzyl alcohol; a rat Achilles tendon treated with a collagenase, 10 mg/ml compound 175 and benzyl alcohol; a rat Achilles tendon treated with a collagenase, 3 mg/ml compound 175, and Phx; and a rat Achilles tendon treated with a collagenase, 10 mg/ml compound 175, and Phx. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) or sham needle puncture for control in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle or compound 175 topical formulations. 24 hours after the last dose, tendons were isolated, fixed with 10% buffered formalin, sectioned and stained with H&E. Group 1 (sham injection), Group 2 (Collagenase-Vehicle with 0.5% Phx), Group 3 (Collagenase-compound 175 3 mg/ml with 0.5% BA), Group 4 (Collagenase-compound 175 10 mg/ml with 0.5% BA), Group 5 (Collagenase-compound 175 3 mg/ml with 0.5% Phx) and Group 6 (Collagenase-COMPOUND 175 10 mg/ml with 0.5% Phx).



FIG. 9 is a plot of tendon histology scores of a rat Achilles tendon; a rat Achilles tendon treated with a collagenase with a vehicle; a rat Achilles tendon treated with a collagenase, 3 mg/ml compound 175, and benzyl alcohol; a rat Achilles tendon treated with a collagenase, 10 mg/ml compound 175, and benzyl alcohol; a rat Achilles tendon treated with a collagenase, 3 mg/ml compound 175, and Phx; and a rat Achilles tendon treated with a collagenase, 10 mg/ml compound 175, and Phx. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) or sham needle puncture for control in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle or COMPOUND 175 topical formulations. 24 hours after the last dose, tendons were isolated, fixed with 10% buffered formalin, sectioned and stained with H&E. Group 1 (sham injection), Group 2 (Collagenase-Vehicle with 0.5% Phx), Group 3 (Collagenase-compound 175 3 mg/ml with 0.5% BA), Group 4 (Collagenase-compound 175 10 mg/ml with 0.5% BA), Group 5 (Collagenase-compound 175 3 mg/ml with 0.5% Phx) and Group 6 (Collagenase-compound 175 10 mg/ml with 0.5% Phx). Blinded histology scoring of tendon injury and inflammation indicates that compound 175 at both 3 and 10 mg/mL, with either BA or Phx as a preservative, significantly ameliorated collagenase-induced tendinopathy (**p<0.01 and ***p<0.001) by student's t test. For this study, a total of 94 sections were scored for Group 1 and 92-116 sections for Groups 2-6.



FIG. 10 is series of plots of plasma concentrations of KC/GRO in rats vs. the number of days elapsed after administration of a vehicle, 3 mg/ml COMPOUND 175 with benzyl alcohol, 10 mg/ml COMPOUND 175 with benzyl alcohol, 3 mg/ml COMPOUND 175 with Phx, and 10 mg/ml compound 175 with Phx. Tendinopathy in rats was induced by injecting collagenase (50 μl, 10 mg/ml Type IA in PBS, pH 7.4, ˜469 units/mg) in the Achilles tendon. After 1 day, rats were dosed once daily for 21 days via topical application with vehicle control or topical compound 175. Blood was collected at various timepoints and plasma was analyzed for KC/GRO by ELISA. Measurement of inflammatory cytokine KC/GRO in the plasma of rats from Group 2 (Collagenase-Vehicle containing 0.5% Phx), Group 3 (Collagenase-compound 175 3 mg/mL with 0.5% BA), Group 4 (Collagenase-compound 175 10 mg/mL with 0.5% BA), Group 5 (Collagenase-compound 175 3 mg/mL with 0.5% Phx) and Group 6 (Collagenase-compound 175 10 mg/mL with 0.5% Phx). Compound 175 at both 3 and 10 mg/mL dose concentrations, with either BA or Phx as preservative significantly decreased the levels of KC/GRO in plasma compared to vehicle (p<0.05 and p<0.01, student's t test as indicated in Table 5), indicating an anti-inflammatory effect of compound 175. These data were generated from 3 replicates per timepoint per group (n=3).



FIG. 11 is a series of plots of compound 175 concentrations vs. time in Sprague-Dawley rat tendons, plasma, and skin after a topical application of 1 mg/mL compound 175 and benzyl alcohol, 10 mg/mL compound 175 and benzyl alcohol, and 10 mg/mL compound 175.





DETAILED DESCRIPTION

This disclosure features the use of one or more indazole-3-carboxamide compounds or salts or analogs thereof, in the treatment of one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease; and/or for promoting wound healing. The methods include administering to a subject (e.g., a subject in need thereof) a therapeutically effective amount of one or more indazole-3-carboxamide compounds or salts or analogs thereof as described anywhere herein.


Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this disclosure belongs. All patents, applications, published applications, and other publications are incorporated by reference in their entirety. In the event that there is a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.


In this specification and in the claims, the following terms have the meanings as defined. As used herein, “alkyl” means a branched, or straight chain chemical group containing only carbon and hydrogen, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl and pentyl. Alkyl groups can either be unsubstituted or substituted with one or more substituents, e.g., halide, alkoxy, acyloxy, amino, amido, cyano, nitro, hydroxyl, mercapto, carboxy, carbonyl, benzyloxy, aryl, heteroaryl, or other functionality that may be suitably blocked, if necessary for purposes of the invention, with a protecting group. Alkyl groups can be saturated or unsaturated (e.g., containing —C═C— or —C≡C— subunits), at one or several positions. Typically, alkyl groups will comprise 1 to 9 carbon atoms, preferably 1 to 6, more preferably 1 to 4, and most preferably 1 to 2 carbon atoms.


As used herein, “carbocyclyl” means a cyclic ring system containing only carbon atoms in the ring system backbone, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cyclohexenyl. Carbocyclyl may include multiple fused rings. Carbocyclyl may have any degree of saturation provided that at least one ring in the ring system is not aromatic. Carbocyclyl groups can either be unsubstituted or substituted with one or more substituents, e.g., alkyl, halide, alkoxy, acyloxy, amino, amido, cyano, nitro, hydroxyl, mercapto, carboxy, carbonyl, benzyloxy, aryl, heteroaryl, or other functionality that may be suitably blocked, if necessary for purposes of the invention, with a protecting group. Typically, carbocyclyl groups will comprise 3 to 10 carbon atoms, preferably 3 to 6.


As used herein, “lower alkyl” means a subset of alkyl having 1 to 3 carbon atoms, and thus is a hydrocarbon substituent, which is linear, or branched. Examples of lower alkyl include methyl, ethyl, n-propyl and isopropyl. Likewise, radicals using the terminology “lower” refer to radicals preferably with 1 to about 3 carbons in the alkyl portion of the radical.


As used herein, “amido” means a H—CON— or alkyl-CON—, carbocyclyl-CON—, aryl-CON—, heteroaryl-CON— or heterocyclyl-CON group wherein the alkyl, carbocyclyl, aryl or heterocyclyl group is as herein described.


As used herein, “aryl” means an aromatic radical having a single-ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) with only carbon atoms present in the ring backbone. Aryl groups can either be unsubstituted or substituted with one or more substituents, e.g., alkyl, amino, cyano, hydroxyl, lower alkyl, haloalkyl, alkoxy, nitro, halo, mercapto, and other substituents. A preferred carbocyclic aryl is phenyl.


As used herein, the term “heteroaryl” means an aromatic radical having one or more heteroatom(s) (e.g., N, O, or S) in the ring backbone and may include a single ring (e.g., pyridine) or multiple condensed rings (e.g., quinoline). Heteroaryl groups can either be unsubstituted or substituted with one or more substituents, e.g., amino, cyano, hydroxyl, lower alkyl, haloalkyl, alkoxy, nitro, halo, mercapto, and other substituents. Examples of heteroaryl include thienyl, pyridinyl, furyl, oxazolyl, oxadiazolyl, pyrrolyl, imidazolyl, triazolyl, thiodiazolyl, pyrazolyl, isoxazolyl, thiadiazolyl, pyranyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, thiazolyl benzothienyl, benzoxadiazolyl, benzofuranyl, benzimidazolyl, benzotriazolyl, cinnolinyl, indazolyl, indolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, purinyl, thienopyridinyl, pyrido[2,3-d]pyrimidinyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl, quinolinyl, thieno[2,3-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[4,3-c]pyridine, pyrazolo[4,3-b]pyridinyl, tetrazolyl, and others.


In these definitions it is clearly contemplated that substitution on the aryl and heteroaryl rings is within the scope of certain embodiments. Where substitution occurs, the radical is called substituted aryl or substituted heteroaryl. Preferably one to three and more preferably one or two substituents occur on the aryl ring. Though many substituents will be useful, preferred substituents include those commonly found in aryl compounds, such as alkyl, cycloalkyl, hydroxy, alkoxy, cyano, halo, haloalkyl, mercapto and the like.


As used herein, “amide” includes both RNR′CO— (in the case of R=alkyl, alkaminocarbonyl-) and RCONR′— (in the case of R=alkyl, alkyl carbonylamino-).


As used herein, the term “ester” includes both ROCO— (in the case of R=alkyl, alkoxycarbonyl-) and RCOO— (in the case of R=alkyl, alkylcarbonyloxy-).


As used herein, “acyl” means an H—CO— or alkyl-CO—, carbocyclyl-CO—, aryl-CO—, heteroaryl-CO— or heterocyclyl-CO— group wherein the alkyl, carbocyclyl, aryl or heterocyclyl group is as herein described. Preferred acyls contain a lower alkyl. Exemplary alkyl acyl groups include formyl, acetyl, propanoyl, 2-methylpropanoyl, t-butylacetyl, butanoyl and palmitoyl.


As used herein, “halo”, “halide” or “halogen” is a chloro, bromo, fluoro or iodo atom radical. Chloro, bromo and fluoro are preferred halides. Most preferred halide is fluorine.


As used herein, “haloalkyl” means a hydrocarbon substituent, which is linear or branched or cyclic alkyl, alkenyl or alkynyl substituted with chloro, bromo, fluoro or iodo atom(s). Most preferred of these are fluoroalkyls, wherein one or more of the hydrogen atoms have been substituted by fluoro. Preferred haloalkyls are of 1 to about 3 carbons in length, more preferred haloalkyls are 1 to about 2 carbons, and most preferred are 1 carbon in length. The skilled artisan will recognize then that as used herein, “haloalkylene” means a diradical variant of haloalkyl, such diradicals may act as spacers between radicals, other atoms, or between the parent ring and another functional group.


As used herein, “heterocyclyl” means a cyclic ring system comprising at least one heteroatom in the ring system backbone. Heterocyclyls may include multiple fused rings. Heterocyclyls may have any degree of saturation provided that at least one ring in the ring system is not aromatic. Heterocyclyls may be substituted or unsubstituted with one or more substituents, e.g., alkyl, halide, alkoxy, acyloxy, amino, amido, cyano, nitro, hydroxyl, mercapto, carboxy, carbonyl, benzyloxy, aryl, heteroaryl, and other substituents, and are attached to other groups via any available valence, preferably any available carbon or nitrogen. More preferred heterocycles are of 5-7 members. In six membered monocyclic heterocycles, the heteroatom(s) are selected from one up to three of O, N or S, and wherein when the heterocycle is five membered, preferably it has one or two heteroatoms selected from O, N, or S. Examples of heterocyclyl include azirinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, 1,4,2-dithiazolyl, 1,3-benzodioxolyl, dihydroisoindolyl, dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[1,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihydropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-dioxolanyl, isoindolinyl, morpholinyl, thiomorpholinyl, piperazinyl, pyranyl, pyrrolidinyl, tetrahydrofuryl, tetrahydropyridinyl, oxazinyl, thiazinyl, thiinyl, thiazolidinyl, isothiazolidinyl, oxazolidinyl, isoxazolidinyl, piperidinyl, pyrazolidinyl imidazolidinyl, thiomorpholinyl, and others.


As used herein, “substituted amino” means an amino radical which is substituted by one or two alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl groups, wherein the alkyl, aryl, heteroaryl or heterocyclyl are defined as above.


As used herein, “substituted thiol” means RS— group wherein R is an alkyl, an aryl, heteroaryl or a heterocyclyl group, wherein the alkyl, cycloalkyl, aryl, heteroaryl or heterocyclyl are defined as above.


As used herein, “sulfonyl” means an alkylSO2, arylSC2, heteroarylSO2, carbocyclylSO2, or heterocyclyl-SO2 group wherein the alkyl, carbocyclyl, aryl, heteroaryl or heterocyclyl are defined as above.


As used herein, “sulfamide” means an alkyl-N—S(O)2N—, aryl-NS(O)2N—, heteroaryl-NS(O)2N—, carbocyclyl-NS(O)2N or heterocyclyl-NS(O)2N— group wherein the alkyl, carbocyclyl, aryl, heteroaryl or heterocyclyl group is as herein described.


As used herein, “sulfonamido” means an alkyl-S(O)2N—, aryl-S(O)2N—, heteroaryl-S(O)2N—, carbocyclyl-S(O)2N— or heterocyclyl-S(O)2N— group wherein the alkyl, carbocyclyl, aryl, heteroaryl or heterocyclyl group is as herein described.


As used herein, “ureido” means an alkyl-NCON—, aryl-NCON—, heteroaryl-NCON—, carbocyclyl-NCON— or heterocyclyl-NCON— group wherein the alkyl, carbocyclyl, aryl, heteroaryl or heterocyclyl group is as herein described.


As used herein, when two groups are indicated to be “linked” or “bonded” to form a “ring,” it is to be understood that a bond is formed between the two groups and may involve replacement of a hydrogen atom on one or both groups with the bond, thereby forming a carbocyclyl, heterocyclyl, aryl, or heteroaryl ring. The skilled artisan will recognize that such rings can and are readily formed by routine chemical reactions, and it is within the purview of the skilled artisan to both envision such rings and the methods of their formations. Preferred are rings having from 3-7 members, more preferably 5 or 6 members. As used herein the term “ring” or “rings” when formed by the combination of two radicals refers to heterocyclic, carbocyclic, aryl, or heteroaryl rings.


The skilled artisan will recognize that some structures described herein may be resonance forms or tautomers of compounds that may be fairly represented by other chemical structures, even when kinetically; the artisan recognizes that such structures are only a very small portion of a sample of such compound(s). Such compounds are clearly contemplated within the scope of this invention, though such resonance forms or tautomers may not be explicitly represented herein.


The compounds provided herein may encompass various stereochemical forms. The compounds also encompass diastereomers as well as optical isomers, e.g. mixtures of enantiomers including racemic mixtures, as well as individual enantiomers and diastereomers, which arise as a consequence of structural asymmetry in certain compounds. Separation of the individual isomers or selective synthesis of the individual isomers is accomplished by application of various methods which are well known to practitioners in the art. Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound.


The term “administration” or “administering” refers to a method of giving a dosage of a compound or pharmaceutical composition to a vertebrate or invertebrate, including a mammal, a bird, a fish, or an amphibian, where the method is, e.g., orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, ontologically, neuro-otologically, intraocularly, subconjuctivally, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, via wound irrigation, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intrameningeally, via inhalation, via endotracheal or endobronchial instillation, via direct instillation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, epidurally, intratympanically, intracistemally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with a prosthetic devices. The preferred method of administration can vary depending on various factors, e.g., the components of the pharmaceutical composition, the site of the disease, the disease involved, and the severity of the disease.


A “diagnostic” as used herein is a compound, method, system, or device that assists in the identification and characterization of a health or disease state. The diagnostic can be used in standard assays as is known in the art.


The term “mammal” is used in its usual biological sense. Thus, it specifically includes humans, cattle, horses, dogs, and cats, but also includes many other species.


The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, co-solvents, complexing agents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like which are not biologically or otherwise undesirable. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. In addition, various adjuvants such as are commonly used in the art may be included. These and other such compounds are described in the literature, e.g., in the Merck Index, Merck & Company, Rahway, N.J. Considerations for the inclusion of various components in pharmaceutical compositions are described, e.g., in Gilman et al. (Eds.) (2010); Goodman and Gilman's: The Pharmacological Basis of Therapeutics. 12th Ed., The McGraw-Hill Companies.


The term “pharmaceutically acceptable salt” refers to salts that retain the biological effectiveness and properties of the compounds of the preferred embodiments and, which are not biologically or otherwise undesirable. In many cases, the compounds of the preferred embodiments are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like; particularly preferred are the ammonium, potassium, sodium, calcium and magnesium salts. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. Many such salts are known in the art, as described in World Patent Publication 87/05297, Johnston et al., published Sep. 11, 1987 (incorporated by reference herein).


“Solvate” refers to the compound formed by the interaction of a solvent and a Wnt pathway inhibitor, a metabolite, or salt thereof. Suitable solvates are pharmaceutically acceptable solvates including hydrates.


“Subject” as used herein, means a human or a non-human mammal, e.g., a dog, a cat, a mouse, a rat, a cow, a sheep, a pig, a goat, a non-human primate or a bird, e.g., a chicken, as well as any other vertebrate or invertebrate.


By “therapeutically effective amount” or “pharmaceutically effective amount” is one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the compound. “Therapeutically effective amount” is also intended to include one or more of the compounds of Formula (I) in combination with one or more other agents that are effective to inhibit Wnt related diseases and/or conditions. The combination of compounds is preferably a synergistic combination. Synergy, as described, for example, by Chou and Talalay, Advances in Enzyme Regulation (1984), 22, 27-55, occurs when the effect of the compounds when administered in combination is greater than the additive effect of the compounds when administered alone as a single agent. In general, a synergistic effect is most clearly demonstrated at sub-optimal concentrations of the compounds. It will be appreciated that different concentrations may be employed for prophylaxis than for treatment of an active disease. This amount can further depend upon the patient's height, weight, sex, age and medical history.


A therapeutic effect relieves, to some extent, one or more of the symptoms of the disease, and includes curing a disease. “Curing” means that the symptoms of active disease are eliminated. However, certain long-term or permanent effects of the disease may exist even after a cure is obtained (such as extensive tissue damage).


“Treat,” “treatment,” or “treating,” as used herein refers to administering a pharmaceutical composition for therapeutic purposes. The term “therapeutic treatment” refers to administering treatment to a patient already suffering from a disease thus causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.


“Morphea” as used herein refers to a skin condition wherein discolored and/or hardened patches appear on the skin (e.g., one or more outer layers of the skin) resulting from excessive collagen deposition.


“Raynaud's syndrome” as used herein refers to a disease in which certain parts of the body (e.g., fingers and/or toes) feel numb and/or cold in response to various stimuli (e.g., cold temperatures and/or stress) due to arterial narrowing.


“Darier's disease” as used herein refers to an autosomal dominant disorder characterized by the appearance of dark crusty patches on the skin (e.g., keratotic papules, keratosis follicularis or dyskeratosis follicularis) that may contain pus.


“Wound healing” as used herein refers to a process by which skin and/or other bodily tissue repairs itself after experiencing, for example, damage and/or trauma.


“Ichthyosis” as used herein refers to a group of genetic skin disorders characterized by the presence of dry, scaly, cracked, and/or flaky skin.


“Tendinopathy” as used herein refers to a disease or disorder of a tendon characterized by inflammation, deterioration, and/or injury of the tendon and/or tissue contacting, near, or associated with the tendon. Tendinopathy includes, for example, inflammation of the tendon (e.g., tendonitis), non-inflammatory degeneration of, for example, the structure and/or composition of a tendon (e.g., tendinosis), inflammation of the paratenon near or in contact with a tendon (e.g., paratenonitis), micro-trauma to the tendon, and rupture of the tendon (e.g., acute, chronic, partial and/or complete rupture). The term also encompasses tenosynovitis, a tendinopathy of the outer lining of the tendon which occurs in certain tendons such as flexor tendons and the Achilles tendon. Symptoms of tendinopathy include pain at rest, upon palpation of the tendon, and/or with movement of, for example, the tendon, tissue, joint, or bone near or associated with the tendon; joint stiffness; difficulty moving; weakness of the joint or muscles surrounding the tendon; redness of the skin near the tendon; swelling of the tendon and/or of tissue near the tendon; and/or crepitus.


“Tendinosis” as used herein, refers to a non-inflammatory injury to the tendon characterized by intratendinous degeneration of the tendon typically in the form of microtears in the tissue in and around the tendon caused by overuse, leading to an increase in the number of tendon repair cells around the area of damage. Degeneration of the tendon is caused by damage to or disorganization of the collagen fibers, cells, and vascular components of the tendon, which can reduce the tendon's tensile strength and can lead to tendon rupture if not treated.


“Tendinitis” as used herein refers to an inflammatory injury to the tendon, characterized by degeneration like that observed in tendinosis, but also accompanied by inflammation of the tendon, vascular disruption and an inflammatory repair response. Tendinitis is often associated with fibroblastic and myofibroblastic proliferation, as well as hemorrhage and organizing granulation tissue. Generally, tendinitis is referred to by the body part involved, such as Achilles tendinitis (affecting the Achilles tendon), or patellar tendinitis (also known as “jumper's knee,” affecting the patellar tendon), though there are certain exceptions, such as lateral epicondylitis (also known as “tennis elbow,” affecting the Extensor Carpi Radialis Brevis tendon). Symptoms can vary from aches or pains and local stiffness to a burning sensation surrounding the entire joint around the inflamed tendon. In some cases, tendonitis is characterized by swelling, sometimes accompanied by heat and redness; there may also be visible knots surrounding the joint. For many patients, the pain is usually worse during and after activity, and the tendon and joint area can become stiffer the following day as muscles tighten from the movement of the tendon.


“Psoriasis” as used herein refers to an autoimmune disease in which skin cells build up and causes raised, red, scaly patches to appear on the skin.


“Dermatitis” (also known as eczema) as used herein refers to generic inflammation of the skin. Specific types of dermatitis include atopic, contact, nummular, photo-induced, and stasis dermatitis. These diseases are characterized by itchiness, red skin, and a rash.


Compounds


Some embodiments of the present invention include compounds, salts, pharmaceutically acceptable salts or pro-drug thereof of Formula (I):




embedded image


In some embodiments of Formula I, R1, R2 and R4 are independently selected from the group consisting of H, C1-9 alkyl, halide, —N(R10)2, —XR10, CN, —OCF3 and —CF3.


In some embodiments of Formula I, R3 is selected from the group consisting of carbocyclylR6, heterocyclylR6, arylR6 and heteroarylR6.


In some embodiments of Formula I, when R3 is heteroaryl, the heteroaryl is not selected from the group consisting of isoquinoline, 1H-pyrrolo[2,3-c]pyridine and tetrazole.


In some embodiments of Formula I, R5 is selected from the group consisting of —(C1-9 alkyl)ncarbocyclylR7, —(C1-9 alkyl)nheterocyclylR7, —(C1-9 alkyl)narylR7 and —(C1-9 alkyl)nheteroarylR7.


In some embodiments of Formula I, R5 is not 4-pyridylR7 when R1, R2 and R4 are H, R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6, 2-pyridylR6, phenylR6, thiazoleR6, imidazoleR6, pyrimidineR6, oxazoleR6,




embedded image



and R6 and R7 are both H.


In some embodiments of Formula I, R5 is not —(CH2)(3-pyridyl)R7 when R1, R2 and R4 are H, R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6 and thiazoleR6, and R6 and R7 are both H.


In some embodiments of Formula I, R5 is not phenylR7 when R1, R2 and R4 are H, R3 is 4-pyridylR6 and R6 and R7 are both H.


In some embodiments of Formula I, R3 is not 3-pyridylR6 when R1, R2 and R4 are H, R5 is selected from the group consisting of phenylR7,




embedded image



and R6 and R7 are both H.


In some embodiments of Formula I, R3 is not oxazoleR6 when R1, R2 and R4 are H, R is selected from the group consisting of




embedded image



and R6 is H.


In some embodiments of Formula I, R3 is not thiazoleR6 when R1, R2 and R4 are H, R5 is selected from the group consisting of




embedded image



and R6 is H.


In some embodiments of Formula I, each R6 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR10, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8, —(C1-9 alkyl)nheteroarylR8, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9alkyl)nN(R10)2, —(C1-9alkyl)nN(R10)SO2R11 and —SO2R11.


In some embodiments of Formula I, each R7 is 1-5 substituents each selected from the group consisting of H, C1-9 alkyl, halide, amino, —OCF3, —CF3, —CN, —XR10, —(C1-9 alkyl)ncarbocyclylR9, —(C1-9 alkyl)nheterocyclylR9, —(C1-9 alkyl)narylR9, —(C1-9 alkyl)nheteroarylR9, —C(═O)R11, —N(R10)C(═O)R11, —(C1-9alkyl)nN(R10)2, —(C1-9alkyl)nN(R10)SO2R11 and —SO2R11.


In some embodiments of Formula I, each R8 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9alkyl)nN(R12)2, —(C1-9alkyl)nN(R12)SO2R13 and —SO2R13.


In some embodiments of Formula I, each R9 is 1-5 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3—CN, —XR12, —C(═O)R13, —N(R12)C(═O)R13, —(C1-9alkyl)nN(R12)2, —(C1-9alkyl)nN(R12)SO2R13 and —SO2R13.


In some embodiments of Formula I, each R10 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8.


In some embodiments of Formula I, each R11 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclylR8, —(C1-9 alkyl)nheterocyclylR8, —(C1-9 alkyl)narylR8 and —(C1-9 alkyl)nheteroarylR8.


In some embodiments of Formula I, each R12 is independently selected from the group consisting of H, C1-9 alkyl, —(C1-9 alkyl)nN(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9 alkyl)naryl and —(C1-9 alkyl)nheteroaryl.


In some embodiments of Formula I, each R13 is independently selected from the group consisting of C1-9 alkyl, —N(R14)2, —(C1-9 alkyl)ncarbocyclyl, —(C1-9 alkyl)nheterocyclyl, —(C1-9 alkyl)naryl and —(C1-9 alkyl)nheteroaryl.


In some embodiments of Formula I, each R14 is independently selected from the group consisting of H, C1-3 alkyl, carbocyclyl and aryl.


In some embodiments of Formula I, each X is selected from the group consisting of a bond, —O— and —S—.


In some embodiments of Formula I, each n is 0 or 1.


In some embodiments of Formula I, X is O.


In some embodiments of Formula I, R1, R2 and R4 are H.


Some embodiments of the present invention include compounds, salts, pharmaceutically acceptable salts or pro-drug thereof of Formula (Ia):




embedded image


wherein:


R3 is selected from the group consisting of 3-pyridylR6, 5-pyrimidinylR6, and 4-pyridazinylR6;


R5 is selected from the group consisting of -heteroarylR7;


R6 is a substituent selected from the group consisting of —(C1-2 alkyl)heterocyclylR8 and -heterocyclylR8;


R7 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, —NH2, —OCF3, —CF3, —CN, —OR10, —(C1-2 alkyl)heterocyclylR9, -heterocyclylR9, and —SO2R11;


R8 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, and —OR12;


each R9 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3, —CN, and —OR12;


R10 is selected from the group consisting of H and C1-3 alkyl;


R11 is C1-3 alkyl; and


each R12 is independently selected from the group consisting of H and C1-3 alkyl.


Some embodiments of the present invention include compounds, salts, pharmaceutically acceptable salts or pro-drug thereof of Formula (Ia):




embedded image


wherein:


R3 is 3-pyridylR6;


R5 is selected from the group consisting of pyridylR7, -pyrimidinylR7, and -pyridazinylR7;


R6 is —CH2heterocyclylR8;


R7 is 1-2 substituents each independently selected from the group consisting of H, F, methyl, —NH2, —CF3, —CN, —OMe, —SO2Me,




embedded image



and


R8 is 1-2 substituents each independently selected from the group consisting of H and halide.


In some embodiments of Formula Ia, R3 is selected from the group consisting of 3-pyridylR6, 5-pyrimidinylR6, and 4-pyridazinylR6;


In some embodiments of Formula Ia, R5 is selected from the group consisting of -heteroarylR7.


In some embodiments of Formula Ia, R5 is selected from the group consisting of -piperazinylR7, -tetrahydropyranylR7, -piperidinylR7, pyrazolylR7, pyrimidinylR7, pyridazinylR7, benzo[d][1,3]dioxolylR7, 2,3-dihydrobenzo[b][1,4]dioxinylR7, pyrazinylR7, and 3-pyridylR7.


In some embodiments of Formula Ia, R6 is a substituent selected from the group consisting of —(C1-2 alkyl)heterocyclylR8 and -heterocyclylR8.


In some embodiments of Formula Ia, R6 is each R6 is 1-2 substituents each selected from the group consisting of —(C1-2 alkyl)heterocyclylR8, -heterocyclylR8, —(C1-2 alkyl)arylR8, —N(R10)C(═O)R11 and —(C1-2 alkyl)N(R10)2.


In some embodiments of Formula Ia, R7 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, —NH2, —OCF3, —CF3, —CN, —OR10, —(C1-2 alkyl)heterocyclylR9, -heterocyclylR9, and —SO2R11.


In some embodiments of Formula Ia, each R7 is 1-2 substituents each selected from the group consisting of unsubstituted C1-3 alkyl, halide, amino, —OCF3, —CF3, —CN, —OR10, —C(═O)R11, —N(R10)C(═O)R11, —N(R10)2, —(C1-2 alkyl)N(R10)2, and —N(R10)SO2R11.


In some embodiments of Formula Ia, R8 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, and —OR12.


In some embodiments of Formula Ia, each R9 is 1-2 substituents each independently selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3, —CN, and —OR12.


In some embodiments of Formula Ia, R10 is selected from the group consisting of Hand C1-3 alkyl.


In some embodiments of Formula Ia, each R10 is independently selected from the group consisting of H, C1-3 alkyl, —(C1-3 alkyl)N(R14)2 and -arylR8.


In some embodiments of Formula Ia, R11 is C1-3 alkyl.


In some embodiments of Formula Ia, R11 is each R11 is independently selected from the group consisting of C1-3 alkyl, —N(R14)2, -carbocyclylR8 and -heterocyclylR8.


In some embodiments of Formula Ia, each R12 is independently selected from the group consisting of H and C1-3 alkyl.


In some embodiments of Formula Ia, R3 is selected from the group consisting of arylR6 and heteroarylR6.


In some embodiments of Formula Ia, when R3 is heteroaryl, the heteroaryl is not selected from the group consisting of isoquinoline, 1H-pyrrolo[2,3-c]pyridine and tetrazole.


In some embodiments of Formula Ia, R5 is selected from the group consisting of -carbocyclylR7, -heterocyclylR7, -arylR7, -heteroarylR7, and —(C1-2 alkyl)heteroarylR7.


In some embodiments of Formula Ia, R5 is not 4-pyridylR7 when R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6, 2-pyridylR6, phenylR6, thiazoleR6, imidazoleR6, pyrimidineR6, oxazoleR6,




embedded image



and R6 and R7 are both H.


In some embodiments of Formula Ia, R5 is not —(CH2)(3-pyridyl)R7 when R3 is selected from the group consisting of 3-pyridylR6, 4-pyridylR6 and thiazoleR6, and R6 and R7 are both H.


In some embodiments of Formula Ia, R5 is not phenylR7 when R3 is 4-pyridylR6 and R6 and R7 are both H.


In some embodiments of Formula Ia, R3 is not 3-pyridylR6 when R5 is selected from the group consisting of phenylR7,




embedded image



and R6 and R7 are both H.


In some embodiments of Formula Ia, R3 is not oxazoleR6 when R5 is selected from the group consisting of




embedded image



and R6 is H.


In some embodiments of Formula Ia, R3 is not thiazoleR6 when R5 is selected from the group consisting of




embedded image



and R6 is H.


In some embodiments of Formula Ia, each R6 is 1-2 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3, —CN, —OR10, —(C1-2 alkyl)heterocyclylR8, -heterocyclylR8, —(C1-2 alkyl)arylR8, —C(═O)R11, —N(R10)C(═O)R11 and —(C1-2 alkyl)N(R10)2.


In some embodiments of Formula Ia, each R7 is 1-2 substituents each selected from the group consisting of H, C1-3 alkyl, halide, amino, —OCF3, —CF3, —CN, —OR10, —(C1-2 alkyl)heterocyclylR9, -heterocyclylR9, -arylR9, —(C1-2 alkyl)arylR9, —C(═O)R11, —N(R10)C(═O)R11, —N(R10)2, —(C1-2alkyl)N(R10)2, —N(R10)SO2R11 and —SO2R11.


In some embodiments of Formula Ia, each R8 is 1-2 substituents each selected from the group consisting of H, C1-2 alkyl, halide, amino, OCF3, —CF3—CN and —OR12.


In some embodiments of Formula Ia, each R9 is 1-2 substituents each selected from the group consisting of H, C1-2 alkyl, halide, amino, —OCF3, —CF3—CN and —OR12.


In some embodiments of Formula Ia, each R10 is independently selected from the group consisting of H, C1-3 alkyl, —(C1-3 alkyl)N(R14)2 and -arylR8.


In some embodiments of Formula Ia, each R11 is independently selected from the group consisting of C1-3 alkyl, —N(R14)2, -carbocyclylR8 and -heterocyclylR8.


In some embodiments of Formula Ia, each R12 is independently selected from the group consisting of H and C1-3 alkyl.


In some embodiments of Formula Ia, each R14 is independently selected from the group consisting of H, C1-3 alkyl and carbocyclyl.


In some embodiments of Formula I or Formula Ia, halide is fluorine.


In some embodiments of Formula I or Formula Ia, R3 is -arylR6.


In some embodiments of Formula I or Formula Ia, R3 is -heteroarylR6.


In some embodiments of Formula I or Formula Ia, R5 is -arylR7.


In some embodiments of Formula I or Formula Ia, R5 is -heteroarylR7.


In some embodiments of Formula I or Formula Ia, R5 is -heterocyclylR7.


In some embodiments of Formula I or Formula Ia, R3 is -heteroarylR6 and R5 is -heteroarylR7.


In some embodiments of Formula I or Formula Ia, R3 is -phenylR6 and R5 is -heteroarylR7.


In some embodiments of Formula I or Formula Ia, R3 is -heteroarylR6 and R5 is -phenylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is —CH2-3-pyridylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -pyridazinylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -pyrazinylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -pyrimidinylR7.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is benzo[d][1,3]dioxolyl.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is 2,3-dihydrobenzo[b][1,4]dioxinyl.


In some embodiments of Formula I or Formula Ia, the aryl is phenyl.


In some embodiments of Formula I or Formula Ia, when R3 is heteroaryl, the heteroaryl is 3-pyridyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is 3-pyridyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is 5-pyrimidinyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is 4-pyridazinyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is pyrazolyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is benzo[d][1,3]dioxolyl.


In some embodiments of Formula I or Formula Ia, when R5 is heteroaryl, the heteroaryl is 2,3-dihydrobenzo[b][1,4]dioxinyl.


In some embodiments of Formula I or Formula Ia, R6 is a heterocyclyl. For example, the heterocyclyl can be selected from the group consisting of morpholinyl, piperazinyl, piperidinyl, tetrahydropyranyl, azetidinyl and pyrrolidinyl. In certain embodiments, R6 is morpholinyl. In another embodiment, R6 is piperazinyl. In another embodiment, R6 is piperidinyl. In another embodiment, R6 is pyrrolidinyl.


In some embodiments of Formula I or Formula Ia, R7 is a heterocyclyl. For example, the heterocyclyl can be selected from the group consisting of morpholinyl, piperazinyl, piperidinyl, tetrahydropyranyl, azetidinyl and pyrrolidinyl. In certain embodiments, R7 is morpholinyl. In another embodiment, R7 is piperazinyl. In another embodiment, R7 is piperidinyl. In another embodiment, R7 is pyrrolidinyl. In another embodiment, R7 is azetidinyl.


In some embodiments of Formula I or Formula Ia, R10 is a carbocyclyl. For example, the carbocyclyl can be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In certain embodiments, R10 is cyclopropyl. In another embodiment, R10 is cyclobutyl. In another embodiment, R10 is cyclopentyl. In another embodiment, R10 is cyclohexyl.


In some embodiments of Formula I or Formula Ia, R11 is a heterocyclyl. For example, the heterocyclyl can be selected from the group consisting of morpholinyl, piperazinyl, piperidinyl, tetrahydropyranyl, azetidinyl and pyrrolidinyl. In certain embodiments, R11 is morpholinyl. In another embodiment, R11 is piperazinyl. In another embodiment, R11 is piperidinyl. In another embodiment, R11 is pyrrolidinyl. In another embodiment, R11 is azetidinyl.


In some embodiments of Formula I or Formula Ia, R11 is a carbocyclyl. For example, the carbocyclyl can be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In certain embodiments, R11 is cyclopropyl. In another embodiment, R11 is cyclobutyl. In another embodiment, R11 is cyclopentyl. In another embodiment, R11 is cyclohexyl.


In some embodiments of Formula I or Formula Ia, R12 is a carbocyclyl. For example, the carbocyclyl can be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In certain embodiments, R12 is cyclopropyl. In another embodiment, R12 is cyclobutyl. In another embodiment, R12 is cyclopentyl. In another embodiment, R12 is cyclohexyl.


In some embodiments of Formula I or Formula Ia, R13 is a heterocyclyl. For example, the heterocyclyl can be selected from the group consisting of morpholinyl, piperazinyl, piperidinyl, tetrahydropyranyl, azetidinyl and pyrrolidinyl. In certain embodiments, R13 is morpholinyl. In another embodiment, R13 is piperazinyl. In another embodiment, R13 is piperidinyl. In another embodiment, R13 is pyrrolidinyl. In another embodiment, R13 is azetidinyl.


In some embodiments of Formula I or Formula Ia, R13 is a carbocyclyl. For example, the carbocyclyl can be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In certain embodiments, R13 is cyclopropyl. In another embodiment, R13 is cyclobutyl. In another embodiment, R13 is cyclopentyl. In another embodiment, R13 is cyclohexyl.


In some embodiments of Formula I or Formula Ia, R6 is one substituent.


In some embodiments of Formula I or Formula Ia, R6 is 1-2 substituents.


In some embodiments of Formula I, R6 is 1-3 substituents.


In some embodiments of Formula I, R6 is 1-4 substituents.


In some embodiments of Formula I or Formula Ia, R6 is H.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is a halide.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —NH2.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —OCF3.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —OCH3.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —CF3.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is -heterocyclylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)heterocyclylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)pyrrolidinylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)pyrrolidinylR8 where R8 is two substituents and both substituents are halides.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)piperidinylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)phenylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is -phenoxyR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is




embedded image


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)2.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)2 where each R10 is independently selected from C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)N(R10)2.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —(CH2)N(R10)2 where each R10 is independently selected from C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)SO2R11.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)C(═O)R11.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)C(═O)R11 where R11 is a heterocyclyl.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is —N(R10)C(═O)R11 where R11 is a carbocyclyl.


In some embodiments of Formula I, R6 is two substituents and the substituents are fluorine and —(C1-9 alkyl)nheterocyclylR8.


In some embodiments of Formula Ia, R6 is two substituents and the substituents are fluorine and -heterocyclylR8.


In some embodiments of Formula Ia, R6 is two substituents and the substituents are fluorine and —(C1-2 alkyl)heterocyclylR8.


In some embodiments of Formula I or Formula Ia, R6 is one substituent and the substituent is select from the group consisting of




embedded image


In some embodiments of Formula I or Formula Ia, R7 is one substituent.


In some embodiments of Formula I or Formula Ia, R7 is 1-2 substituents.


In some embodiments of Formula I, R7 is 1-3 substituents.


In some embodiments of Formula I, R7 is 1-4 substituents.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is a halide.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —NH2.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —OH.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —CF3.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —CN.


In some embodiments of Formula I, R7 is one substituent and the substituent is —XR10 where X is O and R10 is C1-3 alkyl.


In some embodiments of Formula Ia, R7 is one substituent and the substituent is —OR10 and R10 is C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is -phenylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —(CH2)N(R10)2.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —(CH2)N(R10)2 where each R10 is independently selected from C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —(CH2)heterocyclylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —(CH2)pyrrolidinylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is -heterocyclylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is -phenoxyR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —(CH2)phenylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is -phenylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —N(R10)C(═O)R11.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —N(R10)C(═O)R11 where R11 is a carbocyclyl.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —N(R10)2.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —C(═O)R11 where R11 is select from the group consisting of -heterocyclylR8 and —N(R10)2.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —SO2R11.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is —SO2R11; and R11 is C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R7 is two substituents and the substituents are C1-3 alkyl and -heterocyclylR9.


In some embodiments of Formula I or Formula Ia, R7 is one substituent and the substituent is select from the group consisting of




embedded image


embedded image


In some embodiments of Formula I or Formula Ia, R8 is one substituent.


In some embodiments of Formula I or Formula Ia, R8 is 1-2 substituents.


In some embodiments of Formula I, R8 is 1-3 substituents.


In some embodiments of Formula I, R8 is 1-4 substituents.


In some embodiments of Formula I or Formula Ia, R8 is H.


In some embodiments of Formula I or Formula Ia, R8 is one substituent and the substituent is C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R8 is one substituent and the substituent is —OH.


In some embodiments of Formula I or Formula Ia, R8 is one substituent and the substituent is a halide.


In some embodiments of Formula I or Formula Ia, R8 is two substituents and the substituents are halides.


In some embodiments of Formula I, R8 is three substituents and the substituents are halides.


In some embodiments of Formula I or Formula Ia, R9 is one substituent.


In some embodiments of Formula I or Formula Ia, R9 is 1-2 substituents.


In some embodiments of Formula I, R9 is 1-3 substituents.


In some embodiments of Formula I, R9 is 1-4 substituents.


In some embodiments of Formula I or Formula Ia, R9 is H.


In some embodiments of Formula I or Formula Ia, R9 is one substituent and the substituent is C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R9 is one substituent and the substituent is —OH.


In some embodiments of Formula I or Formula Ia, R9 is one substituent and the substituent is a halide.


In some embodiments of Formula I or Formula Ia, R9 is two substituents and the substituents are halides.


In some embodiments of Formula I or Formula Ia, R8 is a —C1-3 alkyl. For example, the —C1-3 alkyl can be selected from the group consisting of methyl, ethyl, n-propyl and isopropyl. In certain embodiments, R8 is methyl. In another embodiment, R8 is ethyl.


In some embodiments of Formula I or Formula Ia, R10 is a —C1-3 alkyl. For example, the —C1-3 alkyl can be selected from the group consisting of methyl, ethyl, n-propyl and iso-propyl. In certain embodiments, R10 is methyl. In another embodiment, R10 is ethyl. In another embodiment, R10 is n-propyl. In another embodiment, R10 is iso-propyl.


In some embodiments of Formula I or Formula Ia, R11 is a —C1-3 alkyl. For example, the —C1-3 alkyl can be selected from the group consisting of methyl, ethyl, n-propyl and iso-propyl. In certain embodiments, R11 is methyl. In another embodiment, R11 is ethyl. In another embodiment, R11 is n-propyl. In another embodiment, R11 is iso-propyl.


In some embodiments of Formula I or Formula Ia, R14 is a —C1-3 alkyl. For example, the —C1-3 alkyl can be selected from the group consisting of methyl, ethyl, n-propyl and iso-propyl. In certain embodiments, R14 is methyl. In another embodiment, R14 is ethyl. In another embodiment, R14 is n-propyl. In another embodiment, R14 is iso-propyl.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)N(R10)2; and R7 is one substituent consisting of —CF3; and each R10 is —C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 and R8 are both H; and the heterocycle is a 5-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 and R8 are both H; and the heterocycle is a 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of CN; R8 is H; and the heterocycle is a 5-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of CN; R8 is H; and the heterocycle is a 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of CF3; R8 is H; and the heterocycle is a 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of -heterocyclylR8; each R8 is H; and the heterocycles are independently selected from a 5 or 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)N(R10)2; and R7 is one substituent consisting of —CN; and each R10 is —C1-3 alkyl.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)N(R10)2; and R7 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R8 is H; each R10 is —C1-3 alkyl; and the heterocycle is a 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of -heterocyclylR8; each R8 is one substituent independently selected from H and —OH; and the heterocycles are independently selected from a 5 or 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of —C(═O)R11; R11 is -heterocyclylR8; each R8 is H; and the heterocycles are independently selected from a 5 or 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of -heterocyclylR8; each R8 is 1-3 substituents independently selected from H and F with the proviso that at least one substituent on one heterocycle is fluorine; and each heterocycle is a 5-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of —C(═O)R11; R11 is —NHR10; R10 is heterocyclylR8; each R8 is H; and the heterocycles are independently selected from a 5 or 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8; R7 is one substituent consisting of —SO2R11; R8 is H; R11 is —C1-3 alkyl; and the heterocycle is a 6-member ring.


In some embodiments of Formula I or Formula Ia, R3 is -3-pyridylR6 and R5 is -3-pyridylR7; R6 is one substituent consisting of —(C1-2 alkyl)heterocyclylR8: R7 is H; R8 is 1-4 substituents independently selected from H and F with the proviso that at least one substituent is fluorine; and the heterocycle is a 5-member ring.


Illustrative compounds of Formula (I) are shown in Table 1.










TABLE 1









embedded image


1







embedded image


2







embedded image


3







embedded image


4







embedded image


5







embedded image


6







embedded image


7







embedded image


8







embedded image


9







embedded image


10







embedded image


11







embedded image


12







embedded image


13







embedded image


14







embedded image


15







embedded image


16







embedded image


17







embedded image


18







embedded image


19







embedded image


20







embedded image


21







embedded image


22







embedded image


23







embedded image


24







embedded image


25







embedded image


26







embedded image


27







embedded image


28







embedded image


29







embedded image


30







embedded image


31







embedded image


32







embedded image


33







embedded image


34







embedded image


35







embedded image


36







embedded image


37







embedded image


38







embedded image


39







embedded image


40







embedded image


41







embedded image


42







embedded image


43







embedded image


44







embedded image


45







embedded image


46







embedded image


47







embedded image


48







embedded image


49







embedded image


50







embedded image


51







embedded image


52







embedded image


53







embedded image


54







embedded image


55







embedded image


56







embedded image


57







embedded image


58







embedded image


59







embedded image


60







embedded image


61







embedded image


62







embedded image


63







embedded image


64







embedded image


65







embedded image


66







embedded image


67







embedded image


68







embedded image


69







embedded image


70







embedded image


71







embedded image


72







embedded image


73







embedded image


74







embedded image


75







embedded image


76







embedded image


77







embedded image


78







embedded image


79







embedded image


80







embedded image


81







embedded image


82







embedded image


83







embedded image


84







embedded image


85







embedded image


86







embedded image


87







embedded image


88







embedded image


89







embedded image


90







embedded image


91







embedded image


92







embedded image


93







embedded image


94







embedded image


95







embedded image


96







embedded image


97







embedded image


98







embedded image


99







embedded image


100







embedded image


101







embedded image


102







embedded image


103







embedded image


104







embedded image


105







embedded image


106







embedded image


107







embedded image


108







embedded image


109







embedded image


110







embedded image


111







embedded image


112







embedded image


113







embedded image


114







embedded image


115







embedded image


116







embedded image


117







embedded image


118







embedded image


119







embedded image


120







embedded image


121







embedded image


122







embedded image


123







embedded image


124







embedded image


125







embedded image


126







embedded image


127







embedded image


128







embedded image


129







embedded image


130







embedded image


131







embedded image


132







embedded image


133







embedded image


134







embedded image


135







embedded image


136







embedded image


137







embedded image


138







embedded image


139







embedded image


140







embedded image


141







embedded image


142







embedded image


143







embedded image


144







embedded image


145







embedded image


146







embedded image


147







embedded image


148







embedded image


149







embedded image


150







embedded image


151







embedded image


152







embedded image


153







embedded image


154







embedded image


155







embedded image


156







embedded image


157







embedded image


158







embedded image


159







embedded image


160







embedded image


161







embedded image


162







embedded image


163







embedded image


164







embedded image


165







embedded image


166







embedded image


167







embedded image


168







embedded image


169







embedded image


170







embedded image


171







embedded image


172







embedded image


173







embedded image


174







embedded image


175







embedded image


176







embedded image


177







embedded image


178







embedded image


179







embedded image


180







embedded image


181







embedded image


182







embedded image


183







embedded image


184







embedded image


185







embedded image


186







embedded image


187







embedded image


188







embedded image


189







embedded image


190







embedded image


191







embedded image


192







embedded image


193







embedded image


194







embedded image


195







embedded image


196







embedded image


197







embedded image


198







embedded image


199







embedded image


200







embedded image


201







embedded image


202







embedded image


203







embedded image


204







embedded image


205







embedded image


206







embedded image


207







embedded image


208







embedded image


209







embedded image


210







embedded image


211







embedded image


212







embedded image


213







embedded image


214







embedded image


215







embedded image


216







embedded image


217







embedded image


218







embedded image


219







embedded image


220







embedded image


221







embedded image


222







embedded image


223







embedded image


224







embedded image


225







embedded image


226







embedded image


227







embedded image


228







embedded image


229







embedded image


230







embedded image


231







embedded image


232







embedded image


233







embedded image


234







embedded image


235







embedded image


236







embedded image


237







embedded image


238







embedded image


239







embedded image


240







embedded image


241







embedded image


242







embedded image


243







embedded image


244







embedded image


245







embedded image


246







embedded image


247







embedded image


248







embedded image


249







embedded image


250







embedded image


251







embedded image


252







embedded image


253







embedded image


254







embedded image


255







embedded image


256







embedded image


257







embedded image


258







embedded image


259







embedded image


260







embedded image


261







embedded image


262







embedded image


263







embedded image


264







embedded image


265







embedded image


266







embedded image


267







embedded image


268







embedded image


269







embedded image


270







embedded image


271







embedded image


272







embedded image


273







embedded image


274







embedded image


275







embedded image


276







embedded image


277







embedded image


278







embedded image


279







embedded image


280







embedded image


281







embedded image


282







embedded image


283







embedded image


284







embedded image


285







embedded image


286







embedded image


287







embedded image


288







embedded image


289







embedded image


290







embedded image


291







embedded image


292







embedded image


293







embedded image


294







embedded image


295







embedded image


296







embedded image


297







embedded image


298







embedded image


299







embedded image


300







embedded image


301







embedded image


302







embedded image


303







embedded image


304







embedded image


305







embedded image


306







embedded image


307







embedded image


308







embedded image


309







embedded image


310







embedded image


311







embedded image


312







embedded image


313







embedded image


314







embedded image


315







embedded image


316







embedded image


317







embedded image


318







embedded image


319







embedded image


320







embedded image


321







embedded image


322







embedded image


323







embedded image


324







embedded image


325







embedded image


326







embedded image


327







embedded image


328







embedded image


329







embedded image


330







embedded image


331







embedded image


332







embedded image


333







embedded image


334







embedded image


335







embedded image


336







embedded image


337







embedded image


338







embedded image


339







embedded image


340







embedded image


341







embedded image


342







embedded image


343







embedded image


344







embedded image


345







embedded image


346







embedded image


347







embedded image


348







embedded image


349







embedded image


350







embedded image


351







embedded image


352







embedded image


353







embedded image


354







embedded image


355







embedded image


356







embedded image


357







embedded image


358







embedded image


359







embedded image


360







embedded image


361







embedded image


362







embedded image


363







embedded image


364







embedded image


365







embedded image


366







embedded image


367







embedded image


368







embedded image


369







embedded image


370







embedded image


371







embedded image


372







embedded image


373







embedded image


374







embedded image


375







embedded image


376







embedded image


377







embedded image


378







embedded image


379







embedded image


380







embedded image


381







embedded image


382







embedded image


383







embedded image


384







embedded image


385







embedded image


386







embedded image


387







embedded image


388







embedded image


389







embedded image


390







embedded image


391







embedded image


392







embedded image


393







embedded image


394







embedded image


395







embedded image


396







embedded image


397







embedded image


398







embedded image


399







embedded image


400







embedded image


401







embedded image


402







embedded image


403







embedded image


404







embedded image


405







embedded image


406







embedded image


407







embedded image


408







embedded image


409







embedded image


410







embedded image


411







embedded image


412







embedded image


413







embedded image


414







embedded image


415







embedded image


416







embedded image


417







embedded image


418







embedded image


419







embedded image


420







embedded image


421







embedded image


422







embedded image


423







embedded image


424







embedded image


425







embedded image


426







embedded image


427







embedded image


428







embedded image


429







embedded image


430







embedded image


431







embedded image


432







embedded image


433







embedded image


434







embedded image


435







embedded image


436







embedded image


437







embedded image


438







embedded image


439







embedded image


440







embedded image


441







embedded image


442







embedded image


443







embedded image


444







embedded image


445







embedded image


446







embedded image


447







embedded image


448







embedded image


449







embedded image


450







embedded image


451







embedded image


452







embedded image


453







embedded image


454







embedded image


455







embedded image


456







embedded image


457







embedded image


458







embedded image


459







embedded image


460







embedded image


461







embedded image


462







embedded image


463







embedded image


464







embedded image


465







embedded image


466







embedded image


467







embedded image


468







embedded image


469







embedded image


470







embedded image


471







embedded image


472







embedded image


473







embedded image


474







embedded image


475







embedded image


476







embedded image


477







embedded image


478







embedded image


479







embedded image


480







embedded image


481







embedded image


482







embedded image


483







embedded image


484







embedded image


485







embedded image


486







embedded image


487







embedded image


488







embedded image


489







embedded image


490







embedded image


491







embedded image


492







embedded image


493







embedded image


494







embedded image


495







embedded image


496







embedded image


497







embedded image


498







embedded image


499







embedded image


500







embedded image


501







embedded image


502







embedded image


503







embedded image


504







embedded image


505







embedded image


506







embedded image


507







embedded image


508







embedded image


509







embedded image


510







embedded image


511







embedded image


512







embedded image


513







embedded image


514







embedded image


515







embedded image


516







embedded image


517







embedded image


518







embedded image


519







embedded image


520







embedded image


521







embedded image


522







embedded image


523







embedded image


524







embedded image


525







embedded image


526







embedded image


527







embedded image


528







embedded image


529







embedded image


530







embedded image


531







embedded image


532







embedded image


533







embedded image


534







embedded image


535







embedded image


536







embedded image


537







embedded image


538







embedded image


539







embedded image


540







embedded image


541







embedded image


542







embedded image


543







embedded image


544







embedded image


545







embedded image


546







embedded image


547







embedded image


548







embedded image


549







embedded image


550







embedded image


551







embedded image


552







embedded image


553







embedded image


554







embedded image


555







embedded image


556







embedded image


557







embedded image


558







embedded image


559







embedded image


560







embedded image


561







embedded image


562







embedded image


563







embedded image


564







embedded image


565







embedded image


566







embedded image


567







embedded image


568







embedded image


569







embedded image


570







embedded image


571







embedded image


572







embedded image


573







embedded image


574







embedded image


575







embedded image


576







embedded image


577







embedded image


578







embedded image


579







embedded image


580







embedded image


581







embedded image


582







embedded image


583







embedded image


584







embedded image


585







embedded image


586







embedded image


587







embedded image


588







embedded image


589







embedded image


590







embedded image


591







embedded image


592







embedded image


593







embedded image


594







embedded image


595







embedded image


596







embedded image


597







embedded image


598







embedded image


599







embedded image


600







embedded image


601







embedded image


602







embedded image


603







embedded image


604







embedded image


605







embedded image


606







embedded image


607







embedded image


608







embedded image


609







embedded image


610







embedded image


611







embedded image


612







embedded image


613







embedded image


614







embedded image


615







embedded image


616







embedded image


617







embedded image


618







embedded image


619







embedded image


620







embedded image


621







embedded image


622







embedded image


623







embedded image


624







embedded image


625







embedded image


626







embedded image


627







embedded image


628







embedded image


629







embedded image


630







embedded image


631







embedded image


632







embedded image


633







embedded image


634







embedded image


635







embedded image


636







embedded image


637







embedded image


638







embedded image


639







embedded image


640







embedded image


641







embedded image


642







embedded image


643







embedded image


644







embedded image


645







embedded image


646







embedded image


647







embedded image


648







embedded image


649







embedded image


650







embedded image


651







embedded image


652







embedded image


653







embedded image


654







embedded image


655







embedded image


656







embedded image


657







embedded image


658







embedded image


659







embedded image


660







embedded image


661







embedded image


662







embedded image


663







embedded image


664







embedded image


665







embedded image


666







embedded image


667







embedded image


668







embedded image


669







embedded image


670







embedded image


671







embedded image


672







embedded image


673







embedded image


674







embedded image


675







embedded image


676







embedded image


677







embedded image


678







embedded image


679







embedded image


680







embedded image


681







embedded image


682







embedded image


683







embedded image


684







embedded image


685







embedded image


686







embedded image


687







embedded image


688







embedded image


689







embedded image


690







embedded image


691







embedded image


692







embedded image


693







embedded image


694







embedded image


695







embedded image


696







embedded image


697







embedded image


698







embedded image


699







embedded image


700







embedded image


701







embedded image


702







embedded image


703







embedded image


704







embedded image


705







embedded image


706







embedded image


707







embedded image


708







embedded image


709







embedded image


710







embedded image


711







embedded image


712







embedded image


713







embedded image


714







embedded image


715







embedded image


716







embedded image


717







embedded image


718







embedded image


719







embedded image


720







embedded image


721







embedded image


722







embedded image


723







embedded image


724







embedded image


725







embedded image


726







embedded image


727







embedded image


728







embedded image


729







embedded image


730







embedded image


731







embedded image


732







embedded image


733







embedded image


734







embedded image


735







embedded image


736







embedded image


737







embedded image


738







embedded image


739







embedded image


740







embedded image


741







embedded image


742







embedded image


743







embedded image


744







embedded image


745







embedded image


746







embedded image


747







embedded image


748







embedded image


749







embedded image


750







embedded image


751







embedded image


752







embedded image


753







embedded image


754







embedded image


755







embedded image


756







embedded image


757







embedded image


758







embedded image


759







embedded image


760







embedded image


761







embedded image


762







embedded image


763







embedded image


764







embedded image


765







embedded image


766







embedded image


767







embedded image


768







embedded image


769







embedded image


770







embedded image


771







embedded image


772







embedded image


773







embedded image


774







embedded image


775







embedded image


776







embedded image


777







embedded image


778







embedded image


779







embedded image


780







embedded image


781







embedded image


782







embedded image


783







embedded image


784







embedded image


785







embedded image


786







embedded image


787







embedded image


788







embedded image


789







embedded image


790







embedded image


791







embedded image


792







embedded image


793







embedded image


794







embedded image


795







embedded image


796







embedded image


797







embedded image


798







embedded image


799







embedded image


800







embedded image


801







embedded image


802







embedded image


803







embedded image


804







embedded image


805







embedded image


806







embedded image


807







embedded image


808







embedded image


809







embedded image


810







embedded image


811







embedded image


812







embedded image


813







embedded image


814







embedded image


815







embedded image


816







embedded image


817







embedded image


818







embedded image


819







embedded image


820







embedded image


821







embedded image


822







embedded image


823







embedded image


824







embedded image


825







embedded image


826







embedded image


827







embedded image


828







embedded image


829







embedded image


830







embedded image


831







embedded image


832







embedded image


833







embedded image


834







embedded image


835







embedded image


836







embedded image


837







embedded image


838







embedded image


839







embedded image


840







embedded image


841







embedded image


842







embedded image


843







embedded image


844







embedded image


845







embedded image


846







embedded image


847







embedded image


848







embedded image


849







embedded image


850









Compound Preparation


The starting materials used in preparing the compounds of the invention are known, made by known methods, or are commercially available. It will be apparent to the skilled artisan that methods for preparing precursors and functionality related to the compounds claimed herein are generally described in the literature. The skilled artisan given the literature and this disclosure is well equipped to prepare any of the compounds.


It is recognized that the skilled artisan in the art of organic chemistry can readily carry out manipulations without further direction, that is, it is well within the scope and practice of the skilled artisan to carry out these manipulations. These include reduction of carbonyl compounds to their corresponding alcohols, oxidations, acylations, aromatic substitutions, both electrophilic and nucleophilic, etherifications, esterification and saponification and the like. These manipulations are discussed in standard texts such as March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure 6th Ed., John Wiley & Sons (2007), Carey and Sundberg, Advanced Organic Chemistry 5th Ed., Springer (2007), Comprehensive Organic Transformations: A Guide to Functional Group Transformations, 2nd Ed., John Wiley & Sons (1999) (incorporated herein by reference in its entirety) and the like.


The skilled artisan will readily appreciate that certain reactions are best carried out when other functionality is masked or protected in the molecule, thus avoiding any undesirable side reactions and/or increasing the yield of the reaction. Often the skilled artisan utilizes protecting groups to accomplish such increased yields or to avoid the undesired reactions. These reactions are found in the literature and are also well within the scope of the skilled artisan. Examples of many of these manipulations can be found for example in T. Greene and P. Wuts Protecting Groups in Organic Synthesis, 4th Ed., John Wiley & Sons (2007), incorporated herein by reference in its entirety.


To further illustrate this invention, the following examples are included. The examples should not, of course, be construed as specifically limiting the invention. Variations of these examples within the scope of the claims are within the purview of one skilled in the art and are considered to fall within the scope of the invention as described, and claimed herein. The reader will recognize that the skilled artisan, armed with the present disclosure, and skill in the art is able to prepare and use the invention without exhaustive examples.


Trademarks used herein are examples only and reflect illustrative materials used at the time of the invention. The skilled artisan will recognize that variations in lot, manufacturing processes, and the like, are expected. Hence the examples, and the trademarks used in them are non-limiting, and they are not intended to be limiting, but are merely an illustration of how a skilled artisan may choose to perform one or more of the embodiments of the invention.


(1H) nuclear magnetic resonance spectra (NMR) were measured in the indicated solvents on a Bruker NMR spectrometer (Avance™ DRX300, 300 MHz for 1H or Avance™ DRX500, 500 MHz for 1H) or Varian NMR spectrometer (Mercury 400BB, 400 MHz for 1H). Peak positions are expressed in parts per million (ppm) downfield from tetramethylsilane. The peak multiplicities are denoted as follows, s, singlet; d, doublet; t, triplet; q, quartet; ABq, AB quartet; quin, quintet; sex, sextet; sep, septet; non, nonet; dd, doublet of doublets; d/ABq, doublet of AB quartet; dt, doublet of triplets; td, triplet of doublets; m, multiplet.


The following abbreviations have the indicated meanings:

    • brine=saturated aqueous sodium chloride
    • CDCl3=deuterated chloroform
    • DCE=dichloroethane
    • DCM=dichloromethane
    • DHP=dihydropyran
    • DIPEA=diisopropylethylamine
    • DMF=N,N-dimethylformamide
    • DMSO-d6=deuterated dimethylsulfoxide
    • ESIMS=electron spray mass spectrometry
    • EtOAc=ethyl acetate
    • EtOH=ethanol
    • h=hour
    • HATU=2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate
    • HCl=hydrochloric acid
    • HOAc=acetic acid
    • H2SO4=sulfuric acid
    • iPrOH=iso-propyl alcohol
    • KOAc=potassium acetate
    • K3PO4=potassium phosphate
    • LAH=lithium aluminum hydride
    • mCPBA=meta-Chloroperoxybenzoic acid
    • MeOH=methanol
    • MgSO4=magnesium sulfate
    • min.=minute
    • MW=microwave
    • NaBH(OAc)3=sodium triacetoxyborohydride
    • NaHCO3=sodium bicarbonate
    • NaHSO3=sodium bisulfite
    • NaHSO4=sodium bisulfate
    • NaOH=sodium hydroxide
    • NH4OH=ammonium hydroxide
    • NMR=nuclear magnetic resonance
    • Pd/C=palladium(O) on carbon
    • PdCl2(dppf)2=1,1′-bis(diphenylphosphino)ferrocene]palladium(II) chloride
    • Pd2(dba)3=tris(dibenzylideneacetone)dipalladium(0)
    • Pd(PPh3)4=tetrakis(triphenylphosphine)palladium(0)
    • PPTS=pyridinium p-toluenesulfonate
    • r.t.=room temperature
    • satd.=saturated
    • soln.=solution
    • Reflx.=heated to reflux
    • TEA=triethylamine
    • TFA=trifluoroacetic acid
    • THF=tetrahydrofuran
    • TLC=thin layer chromatography
    • Tr-Cl=trityl chloride or triphenylmethyl chloride


The following example schemes are provided for the guidance of the reader, and collectively represent an example method for making the compounds provided herein. Furthermore, other methods for preparing compounds of the invention will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. The skilled artisan is thoroughly equipped to prepare these compounds by those methods given the literature and this disclosure. The compound numberings used in the synthetic schemes depicted below are meant for those specific schemes only, and should not be construed as or confused with same numberings in other sections of the application. Unless otherwise indicated, all variables are as defined above.


General Procedures


Compounds of Formula I of the present invention can be prepared as depicted in Scheme 1.




embedded image


Scheme 1 describes a method for preparation of indazole-3-carboxamide derivatives (I) by first forming the Weinreb amide (III) of a 1H-indazole-3-carboxylic acid (II). The Weinreb amide (III) is reacted with (bis(trifluoroacetoxy)iodo)benzene to produce the 5-iodo-1H-indazole-3-carboxylic acid (IV) followed by THF protection of the indazole nitrogen. The Weinreb amide of protected indazole V is reduced to aldehyde VI followed by reaction with bis(pinacolato)diboron to give the pinacol ester (VII). Suzuki coupling with a variety of aromatic and nonaromatic bromides yields the R3 substituted indazole VIII. Oxidation of the aldehyde to the acid (IX) followed by HATU mediated coupling of a variety of amines and sequent deprotection produces the desired indazole-3-carboxamide derivatives (I).


Compounds of Formula I of the present invention can also be prepared as depicted in Scheme 2.




embedded image


Scheme 2 describes an alternative method for preparation of indazole-3-carboxamide derivatives (I) by bromination of the indazole 5-position followed by esterification to form ester XII. The indazole nitrogen is THF protected and the ester is hydrolyzed to acid XIV. The acid is coupled with a variety of amines to produce amide XV which is then coupled with a variety of boronic acids (Route 1) to give X. Alternatively, XV can be converted to the boronate ester and then couple to a variety of bromides (Route 2) to yield X. Final deprotection of the indazole nitrogen yields the desired indazole-3-carboxamide derivatives (I).


Compounds of Formula I of the present invention can also be prepared as depicted in Scheme 3.




embedded image


Scheme 3 describes another alternative method for preparation of indazole-3-carboxamide derivatives (I) by bromination of the indazole 5-position followed by either Route 1: esterification to form ester XII, then trityl protection of the indazole nitrogen and then finally hydrolyzed of the ester to acid XVII; or Route 2: trityl protection of the indazole nitrogen directly to acid XVII. The acid is coupled with a variety of amines to produce amide XVIII which is then coupled with a variety of boronic acids (Route 3) to give XIX. Alternatively, XVIII can be converted to the boronate ester and then couple to a variety of bromides (Route 4) to yield XIX. Final deprotection of the indazole nitrogen yields the desired indazole-3-carboxamide derivatives (I).


ILLUSTRATIVE COMPOUND EXAMPLES

Preparation of intermediate 3-(5-bromopyridin-3-yl)-1,1-dimethylurea (XXII) is depicted below in Scheme 4.




embedded image



Step 1


3-Amino-5-bromo pyridine (XX) (1.0 g, 5.78 mmol) was dissolved in pyridine and cooled to 0° C. before adding dimethyl carbamyl chloride (XXI) (0.683 g, 6.35 mmol). The reaction mixture was stirred at room temperature for 2 h and then heated overnight at 60° C. under argon. The solution was cooled to room temperature, poured into ice water and extracted with EtOAc. The organic extract was dried over MgSO4, filtered and concentrated to a residue to afford 3-(5-bromopyridin-3-yl)-1,1-dimethylurea (XXII) as a brown solid, (1.24 g, 5.09 mmol, 88% yield). 1H NMR (DMSO-d6) δ ppm 8.67-8.64 (m, 2H), 8.23 (d, J=7.8 Hz, 1H), 2.93 (s, 6H); ESIMS found for C8H10BrN3O m/z 245.05 (M+H).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 4.




embedded image


N-(5-bromopyridin-3-yl)morpholine-4-carboxamide (XXIII): Tan solid (0.82 g, 48%). 1H NMR (DMSO-d6) 3.43-3.45 (m, 4H), 3.60-3.62 (m, 4H), 8.21 (t, J=2.0 Hz, 1H), 8.26 (d, J=2.0 Hz, 1H), 8.62 (d, J=2.2 Hz, 1H), 8.91 (s, 1H); ESIMS found for C10H12BrN3O2 m/z 286 (M+H).




embedded image


N-(5-bromopyridin-3-yl)cyclopropanecarboxamide (XXIV): Off white solid, (83% yield), 1H NMR (CDCl3, 400 MHz) δ ppm 8.46-8.39 (m, 3H), 7.54 (bs, 1H), 1.56-1.50 (m, 1H), 1.13-1.07 (m, 2H), 0.96-0.90 (m, 2H); ESIMS found for C9H9BrN2O m/z 240.85 (M+H).


Preparation of intermediate (XXVI) is depicted below in Scheme 5.




embedded image


To a solution of 5-bromonicotinaldehyde (XXV) (5.0 g, 26.9 mmol) in DCE (108 mL) was added dimethylamine-HCl (4.39 g, 53.8 mmol) and TEA (7.5 g, 53.8 mmol). The reaction was stirred at room temperature for 1 h. NaBH(OAc)3 was added and the reaction was stirred overnight at room temperature. The reaction was diluted with DCM and sat. aq. NaHCO3. The organic layer was separated, washed with water, brine, dried and concentrated under vacuum to produce 1-(5-bromopyridin-3-yl)-N,N-dimethylmethanamine (XXVI) as a brown liquid (92.6% yield). 1H NMR (CDCl3) δ ppm 2.15 (s, 6H), 3.43 (s, 2H), 7.94 (s, 1H), 8.47 (d, J=2 Hz, 1H), 8.59 (d, J=3 Hz, 1H); ESIMS found for C8H11BrN2 m/z 215 (MBr79+H) and 217 (MBr81+H).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 5.




embedded image


3-Bromo-5-(pyrrolidin-1-ylmethyl)pyridine (XXVII): Golden liquid (1.35 g, 97% yield). 1H NMR (DMSO-d6) 1.68-1.71 (m, 4H), 2.42-2.44 (m, 4H), 3.60 (s, 2H), 7.96 (s, 1H), 8.48 (d, J=2 Hz, 1H), 8.58 (d, J=3 Hz. 1H); ESIMS found for C10H13BrN2 m/z 242 (M+H).




embedded image


3-Bromo-5-(piperidin-1-ylmethyl)pyridine (XXVIII): Brown liquid (13.1 g, 94% yield). 1H NMR (DMSO-d6) 1.36-1.39 (m, 2H), 1.46-1.51 (m, 4H), 2.31-2.32 (m, 4H), 3.46 (s, 2H), 7.94 (s, 1H), 8.47 (d, J=2 Hz, 1H), 8.58 (d, J=3 Hz, 1H); ESIMS found for C11H15BrN2 m/z 257 (M+H).




embedded image


4-((5-Bromopyridin-3-yl)methyl)morpholine (XXIX): Brown oil (1.02 g, 35.6% yield). ESIMS found for C10H13BrN2O m/z 258 (M+H).




embedded image


1-((5-Bromopyridin-3-yl)methyl)-4-methylpiperazine (XXX): Brown oil (0.93 g, 64% yield). 1H NMR (DMSO-d6) 2.14 (s, 3H), 2.27-2.37 (m, 8H), 3.49 (s, 2H), 7.95 (s, 1H), 8.47 (d, J=1.7 Hz, 1H), 8.59 (d, J=2.2 Hz. 1H); ESIMS found for C11H16BrN3 m/z 212 (M+H).




embedded image


1-(3-Bromo-5-fluorobenzyl)-4-methylpiperazine (XXXI): Light yellow oil (2.07 g, 68% yield). 1H NMR (DMSO-d6) 2.14 (s, 3H), 2.28-2.40 (m, 8H), 3.46 (s, 2H), 7.15-7.17 (m, 1H), 7.35 (s, 1H), 7.40-7.42 (m, 1H); ESIMS found for C12H16BrFN2 m/z 288 (M+H).




embedded image


1-(5-Bromopyridin-3-yl)piperidin-4-ol (XXXII): Brown oil (2.15 g, 7.93 mmol, 72.7% yield). 1H NMR (DMSO-d6) 1.34-1.41 (m, 2H), 1.67-1.71 (m, 2H), 2.03-2.07 (m, 2H), 2.62-2.64 (m, 2H), 3.42-3.46 (m, 1H), 3.47 (s, 2H), 4.55 (d, J=4.2 Hz, 1H), 7.93-7.94 (m, 1H), 8.46 (d, J=1.6 Hz, 1H), 8.58 (d, J=2.2 Hz, 1H); ESIMS found for C11H15BrN2O m/z 272 (M+H).




embedded image


3-Bromo-5-((3,3-difluoropyrrolidin-1-yl)methyl)pyridine (XXXIII): Brown liquid (7.38 g, 26.64 mmol, 94.9% yield). 1H NMR (DMSO-d6) 2.21-2.30 (m, 2H), 2.70 (t, J=7 Hz, 2H), 2.89 (t, J=13 Hz, 2H), 3.66 (s, 2H), 7.95-7.98 (m, 1H), 8.57 (d, J=1.7 Hz, 1H), 8.61 (d, J=2.2 Hz, 1H); ESIMS found for C10H11BrF2N2 m/z 276 (M+H).


Preparation of 3-benzyl-5-bromopyridine (XXXVI) is depicted below in Scheme 6.




embedded image



Step 1


To a solution of 3,5-dibromopyridine (XXXIV) (1.03 g, 4.36 mmol) in THF (7 mF) under argon was added CuI (50 mg, 0.26 mmol) and PdCl2(dppf)2 (178 mg, 0.22 mmol). Benzylzinc(II) bromide (XXXV) (0.5M in THF) (13.09 mF, 6.55 mmol) was slowly added by syringe. The reaction was heated at 50° C. over the weekend. The reaction was quenched with water and extracted with EtOAc. The EtOAc was separated, washed with water, brine, dried over MgSO4 and concentrated under vacuum. The residue was purified on a silica gel column (100% hexanes→5:95 EtOAc:hexanes) to afford 3-benzyl-5-bromopyridine (XXXVI) (0.614 g, 2.47 mmol, 57% yield) as a light brown oil. 1H NMR (DMSO-d6) δ ppm 3.98 (s, 2H), 7.19-7.23 (m, 1H), 121-132 (m, 4H), 7.92-7.93 (m, 1H), 8.51 (d, J=2 Hz, 1H), 8.54 (d, J=3 Hz, 1H); ESIMS found for C12H10BrN m/z 248 (M+H).


Preparation of 3-bromo-5-phenoxypyridine (XXXIX) is depicted below in Scheme 7.




embedded image



Step 1


To a solution of 3,5-dibromopyridine (XXXVII) (1.00 g, 4.24 mmol) in NMP (11 mF) was added phenol (XXXVIII) (398 mg, 4.24 mmol) and CsCO3 (1.38 g, 4.24 mmol). The reaction was heated at 100° C. over the weekend. The reaction was then partitioned between Et2O/water. The Et2O was separated, washed with 2× water, brine, dried over MgSO4 and concentrated under vacuum. The residue was purified on a silica gel column (100% hexanes→2:98 EtOAc:hexanes) to afford 3-bromo-5-phenoxypyridine (XXXIX) (535 mg, 2.14 mmol, 50% yield) as a clear oil. 1H NMR (DMSO-d6) δ ppm 7.13-7.15 (m, 2H), 7.23-7.26 (m, 1H), 7.43-7.46 (m, 2H), 7.69-7.70 (m, 1H), 8.37 (d, J=3 Hz, 1H), 8.49 (d, J=2 Hz, 1H); ESIMS found for C11H8BrNO m/z 250 (M+H).


Preparation of l-(5-bromopyridin-3-yl)-4-methylpiperazine (XL) is depicted below in Scheme 8.




embedded image



Step 1


To a solution of 3,5-dibromopyridine (XXXVIII) (2.90 g, 12.24 mmol) in dry DMF (20 mL) was added 1-methylpiperazine (2.987 mL, 26.93 mmol) and K2CO3 (5.58 g, 40.39 mmol). The reaction was heated at 120° C. overnight. An additional portion of 1-methylpiperazine (6 mL) was added and heating was continued for another 24 h. The reaction was poured into ice water and filtered. The filtrate was extracted with 66% MeOH/CHCl3. The organic layer was dried over MgSO4, filtered and concentrated under vacuum to yield 1-(5-bromopyridin-3-yl)-4-methylpiperazine (XL) as a brown viscous oil (2.49 g, 9.76 mmol, 79.8% yield). ESIMS found for C10H14BrN3 m/z 256 (M+H).


The following intermediate was prepared in accordance with the procedure described in the above Scheme 8.




embedded image


4-(5-Bromopyridin-3-ylmorpholine (XLI): Yellow solid (1.12 g, 4.61 mmol, 64.9% yield). ESIMS found for C9H11BrN2O m/z 244.1 (M+H).


Preparation of 5-bromo-N-cyclohexylnicotinamide (XLIV) is depicted below in Scheme 9.




embedded image



Step 1


To a solution of 5-bromonicotinic acid (XLII) (500 mg, 2.49 mmol) in DMF (8 mL) was added cyclohexanamine (XLIII) (247 mg, 2.49 mmol) and DIPEA (643 mg, 4.98 mmol). The reaction was cooled at 0° C. before adding HATU (947 mg, 2.49 mmol). The reaction was warmed to room temperature and stirred for 4 hrs. The reaction was diluted with EtOAc, washed with 2× water, brine, dried over MgSO4 and concentrated under vacuum to yield crude 5-bromo-N-cyclohexylnicotinamide (XLIV). The product was used without further purification. ESIMS found for C12H15BrN2O m/z 283 (M+H).


Preparation of 3-bromo-5-(((2R,6S)-2,6-dimethylpiperidin-1-yl)methyl)pyridine (XLVII) is depicted below in Scheme 10.




embedded image



Step 1


To a solution of 5-bromonicotinaldehyde (XXV) (2.05 g, 11.0 mmol) in MeOH (85 mL) was added NaBH, (832 mg, 21.99 mmol). The reaction was stirred at room temperature for 1 h. The reaction was quenched with saturated aqueous NH4Cl (5 mL). The reaction was concentrated under vacuum and the residue was partitioned between saturated aqueous NH4Cl/EtOAc. The organic layer was separated, washed with water, brine, dried over MgSO4 and concentrated under vacuum to yield crude (5-bromopyridin-3-ylmethanol (XLV) as a golden oil (1.54 g, 8.2 mmol, 74% yield). The product was used without further purification. ESIMS found for C6H6BrNO m/z 188 (M+H).


Step 2


(5-Bromopyridin-3-ylmethanol (XLV) (1.54 g, 8.2 mmol) was treated with 4M HCl in dioxane (10 mL) at 0° C. and then evaporated. The residue was dissolved in SOCl2 (4 mL) and refluxed for 2 hrs. The SOCl2 was removed and the residue was triturated with hexane to produce HCl salt of 3-bromo-5-(chloromethyl)pyridine (XLVI) as a brown solid (1.30 g, 5.4 mmol, 66% yield). The product was used without further purification. ESIMS found for C6H5BrClN m/z 206 (M+H).


Step 3


To a solution of 3-bromo-5-(chloromethylpyridine (XLVI) (1.17 g, 4.8 mmol) in MeCN (0.2 mL) and (2S,6R)-2,6-dimethylpiperidine (2.6 mL, 19.3 mmol) was added K2CO3 (667 mg, 4.8 mmol). The reaction was refluxed for 5 hrs. TLC showed the presence of starting material so additional (2S,6R)-2,6-dimethylpiperidine (2.0 mL, 14.8 mmol) was added and the reaction was refluxed for an additional 5 hrs. The solvent was removed and the residue was partitioned between EtOAc/water. The EtOAc was separated and washed with brine, dried over MgSO4 and concentrated under vacuum. The residue was purified on a silica gel column (100% hexanes→6:94 EtOAc:hexanes) to afford 3-bromo-5-(((2R,6S)-2,6-dimethylpiperidin-1-yl)methyl)pyridine (XLVII) as a clear oil (728 mg, 2.57 mmol, 53% yield). 1H NMR (DMSO-d6) δ ppm 0.92 (d, J=8 Hz, 6H), 1.21-1.32 (m, 3H), 1.52-1.55 (m, 2H), 1.59-1.63 (m, 1H), 2.42-2.46 (m, 2H), 3.73 (s, 2H), 7.97-7.98 (m, 1H), 8.50 (d, J=3 Hz, 1H), 8.55-8.56 (m, 1H); ESIMS found for C13H19BrN2 m/z 283 (M+H).


Preparation of intermediate 3′-fluorobiphenyl-3-amine (LI) is depicted below in Scheme 11.




embedded image



Step 1


A 25 mL microwave vessel was charged with 1-bromo-3-nitrobenzene (XLVIII) (0.61 g, 3.0 mmol), 3-fluorophenylboronic acid (XLIX) (0.46 g, 3.3 mmol), potassium phosphate tribasic (0.95 g, 4.5 mmol), 1,4-dioxane (15.0 mL), and water (3.0 mL). Tetrakis(triphenylphosphine)palladium(0) (0.17 g, 0.15 mmol) was added, and the reaction was placed in a microwave reactor for 1 h at 95° C. An additional 3-fluorophenylboronic acid (0.20 g) and tetrakistriphenylphosphine)palladium(0) (0.05 g) were added, and the reaction was heated for another 1 h at 95° C. in a microwave reactor. The organic solvent was separated from the water and concentrated to a residue. The residue was then purified by flash chromatography using a 25 g Thomson normal phase silica gel cartridge (100% hexanes→1:99 EtOAc:hexanes) to afford 3′-fluoro-3-nitrobiphenyl (L) (0.63 g, 2.91 mmol, 97% yield) as a white solid. 1H NMR (DMSO-d6) δ ppm 8.48 (t, J=2.0 Hz, 1H), 8.26-8.24 (m, 1H), 8.20-8.18 (m, 1H), 7.78 (t, J=8 Hz, 1H), 7.70-7.68 (m, 1H), 7.67-7.65 (m, 1H), 7.59-7.56 (m, 1H), 7.32-7.28 (m, 1H).


Step 2


10% Palladium on carbon (0.095 g) was added to a solution of 3′-fluoro-3-nitrobiphenyl (L) (0.63 g, 2.88 mmol) in EtOH (20.0 mL). The flask was evacuated and replaced with a hydrogen atmosphere. The solution was stirred at room temperature for 5 h under hydrogen. The catalyst was filtered through a pad of Celite, and the solvent was removed under reduced pressure. The residue was purified by flash chromatography using a 40 g Thomson normal phase silica gel cartridge (100% hexanes→15:85 EtOAc:hexanes) to afford 3′-fluorobiphenyl-3-amine (LI) (0.34 g, 1.81 mmol, 63% yield) as a light yellow oil. 1H NMR (DMSO-d6) δ ppm 7.47-7.44 (m, 1H), 7.40-7.39 (m, 1H), 7.36-7.33 (m, 1H), 7.15-7.14 (m, 1H), 7.10 (t, J=7.7 Hz, 1H), 6.85-6.84 (m, 1H), 6.80-6.79 (m, 1H), 6.60-6.58 (m, 1H), 5.18 (s, 2H); ESIMS found for C12H10FN m/z 188 (M+H).


Preparation of intermediate 5-(3-fluorophenyl)pyridin-3-amine (LIII) is depicted below in Scheme 12.




embedded image



Step 1


To a microwave vial was added 3-amino-5-bromopyridine (LII) (0.400 g, 2.31 mmol), 3-fluorophenyl boronic acid (XLIX) (0.356 g, 2.54 mmol), tetrakistriphenylphosphine)palladium(0) (0.133 g, 0.116 mmol), potassium phosphate (0.736 g, 3.47 mmol), water (1 mL), and DMF (5 mL). The reaction vial was capped, purged with argon and heated under microwave irradiation for 1 h at 180° C. The solution was filtered through a pad of Celite and concentrated under vacuum. The residue was purified by column chromatography (4:6 EtOAc:hexanes→7:3 EtOAc:hexanes) to afford the 5-(3-fluorophenyl)pyridin-3-amine (LIII) (0.360 g, 1.92 mmol, 83% yield) as a yellow-white solid. ESIMS found for C11H9FN2 m/z 189.1 (M+H).


Preparation of intermediate 5-((dimethylamino)methyl)pyridin-3-amine (LVII) is depicted below in Scheme 13.




embedded image



Step 1


5-Bromonicontinaldehyde (XXV) (5.01 g, 26.9 mmol) and dimethylamine hydrochloride (4.39 g, 53.8 mmol) were suspended in 1,2-dichloroethane (108 mL). Triethylamine (7.50 mL, 53.8 mmol) was added, and the reaction was stirred at room temperature for 1 h. Sodium triacetoxyborohydride (8.56 g, 40.4 mmol) was added, and the reaction was further stirred at room temperature overnight. The reaction was diluted with saturated sodium bicarbonate solution and DCM. The organic layer was separated, washed sequentially with water and brine, dried over MgSO4, filtered and concentrated to give 1-(5-bromopyridin-3-yl)-N,N-dimethylmethanamine (LIV) (1.19 g, 23.9 mmol, 89% yield) as a brown oil: 1H NMR (DMSO-d6) δ ppm 8.59 (d, J=3 Hz, 1H), 8.47 (d, J=2 Hz, 1H), 7.94 (s, 1H), 3.43 (s, 2H), 2.15 (s, 6H); ESIMS found for C8H11BrN2 m/z 215 (M+H).


Step 2


In a 25 mL microwave vessel, l-(5-bromopyridin-3-yl)-N,N-dimethylmethanamine (LIV) (1.27 g, 5.92 mmol), 4-methoxybenzylamine (LV) (0.77 mL, 5.92 mmol), cesium carbonate (2.70 g, 8.29 mmol) and xanthphos (0.17 g, 0.30 mmol) were suspended in xylenes (12.0 mL). The solvent was degassed, and tris(dibenzylideneacetone)dipalladium(0) (0.27 g, 0.30 mmol) was added. The vessel was sealed, and the reaction was heated to 130° C. for 5 h in a microwave reactor. The solvent was decanted away from the solid material and concentrated to a residue. The residue was purified by silica gel chromatography using a 40 g Thomson normal-phase silica gel cartridge (100% CHCl3→3:97 MeOH[7N NH3]:CHCl3) to afford 5-((dimethylamino)methyl)-N-(4-methoxybenzyl)pyridin-3-amine (LVI) (0.68 g, 2.49 mmol, 42% yield) as a yellow solid. 1H NMR (DMSO-d6) δ ppm 7.84 (d, J=3 Hz, 1H), 7.64 (d, J=2 Hz, 1H), 7.27 (d, J=1 Hz, 2H), 6.88 (d, J=11 Hz, 2H), 6.83-6.82 (m, 1H), 6.35 (t, J=8 Hz, 1H), 4.20 (d, J=8 Hz, 2H), 3.72 (s, 3H), 3.24 (s, 2H), 2.08 (s, 6H); ESIMS found for C16H21N3O m/z 212 (M+H).


Step 3


5-((dimethylamino)methyl)-N-(4-methoxybenzyl)pyridin-3-amine (LVI) (0.15 g, 0.56 mmol) was dissolved in TFA (2.0 mL) and stirred at room temperature for 1 h. The TFA was removed, and the residue was treated with 7N ammonia in MeOH/chloroform mixture (7/93) to neutralize the TFA and concentrated again to a residue. The residue was purified by flash silica gel chromatography utilizing a 4 g Thomson normal-phase silica gel cartridge (100% CHCl3→3:97 MeOH[7N NH3]:CHCl3) to afford 5-((dimethylamino)methyl)pyridin-3-amine (LVII) (0.044 g, 0.29 mmol, 52% yield) as a brown oil. ESIMS found for C8H13N3 m/z 152 (M+H).


The following intermediate was prepared in accordance with the procedure described in the above Scheme 13.




embedded image


5-((4-Methylpiperazin-1-yl)methyl)pyridin-3-amine (LVIII): Dark yellow solid (138 mg, 0.67 mmol, 71% yield). ESIMS found for C11H18N4 m/z 207 (M+H).


Preparation of intermediate dipyrrolidin-1-ylmethyl)pyridin-3-amine (LXIII) is depicted below in Scheme 14.




embedded image



Step 1


To a suspension of methyl 5-nitropicolinate (LIX) (1.282 g, 7.03 mmol) in DCM (25 mL) stirred at −78° C. under argon was slowly added DIBAL (1M in toluene) (9.14 mL, 9.14 mmol). The solution was allowed to warm to room temperature over 3 h. An aqueous solution of potassium sodium tartrate was added, diluted further with water and DCM. The solution was stirred at room temperature for another 30 min before the organic layer was separated. The aqueous layer was extracted 2×DCM, combined with the organic layer, dried over MgSO4, filtered and evaporated under reduced pressure. The residue was purified by column chromatography to produce 5-nitropicolinaldehyde (LX) as a brown oil (0.64 g, 4.2 mmol, 60% yield). NMR (DMSO-d6) δ ppm 8.17 (d, J=9 Hz, 1H), 8.81 (dd, J=9 Hz, J=2 Hz, 1H), 9.56 (d, J=2 Hz, 1H), 10.08 (s, 1H).


Step 2


Preparation of 5-nitrosopyrrolidin-1-ylmethyl)pyridine (LXII) was performed following the procedure listed in Scheme 5, Step 1. Purple oil (0.41 g, 1.98 mmol, 86% yield). 1H NMR (DMSO-d6) δ ppm 9.28 (d, J=3 Hz, 1H), 8.56 (dd, J=11 Hz, 3 Hz, 1H), 7.72 (d, J=11 Hz, 1H), 3.85 (s, 2H), 2.53-2.50 (m, 4H), 1.75-1.70 (m, 4H).


Step 3


Preparation of intermediate dipyrrolidin-1-ylmethyl)pyridin-3-amine (LXIII) was performed following the procedure listed in Scheme 11, Step 2. Dark brown oil (0.35 g, 1.97 mmol, quantitative). ESIMS found for C10H15N3 m/z 178 (M+H).


The following intermediate was prepared in accordance with the procedure described in the above Scheme 14.




embedded image


6-((4-Methylpiperazin-1-yl)methyl)pyridin-3-amine (LXIV): Brown oil (120 mg, 0.58 mmol, 100% yield). ESIMS found for C11H18N4 m/z 207 (M+H).


Preparation of intermediate 6-(3-fluorophenoxy)pyridin-3-amine (LXVIII) is depicted below in Scheme 15.




embedded image



Step 1


A solution of 2-chloro-5-nitropyridine (LXV) (1.98 g, 12.5 mmol) and 3-fluorophenol (LXVI) (1.4 g, 12.5 mmol) in pyridine (20 mL) was heated at 120° C. overnight under argon. The solution was cooled to room temperature and concentrated under vacuum. The residue was dissolved in EtOAc, washed with water, brine, dried over MgSO4 and evaporated. The residue was purified by silica gel column chromatography (100% hexane→2:98 EtOAc:hexane) to give 2-(3-fluorophenoxy)-5-nitropyridine (LXVII) as a yellow viscous oil (2.27 g, 9.7 mmol, 77% yield). 1H NMR (DMSO-d6) δ ppm 7.11 (dd, J=8 Hz. J=2 Hz. 1H), 7.17 (dt, J=8 Hz, J=6 Hz. 1H), 7.23 (td, J=10 Hz, J=2 Hz, 1H), 7.31 (d, J=9 Hz, 1H), 7.52 (q. J=9 Hz. 1H), 8.64 (dd, J=9 Hz, J=3 Hz, 1H), 9.05 (d, J=3 Hz, 1H); ESIMS found for C11H7FN2O3 m/z 234.9 (M+H).


Step 2


Preparation of intermediate 6-(3-fluorophenoxy)pyridin-3-amine (LXVIII) was performed following the procedure listed in Scheme 11, Step 2. Black green viscous oil (1.90 g, 9.3 mmol, 96% yield). 1H NMR (DMSO-d6) δ ppm 5.18 (brs, 2H), 6.74-6.83 (m, 3H), 6.90 (dt, 1H), 7.09 (dd, J=9 Hz, J=3 Hz, 1H), 7.34 (q, J=7 Hz, 1H), 7.57 (d, J=3 Hz, 1H); ESIMS found for C11H9FN2O m/z 204.4 (M+).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 15.




embedded image


6-(4-Fluorophenoxy)pyridin-3-amine (LXIX): Dark brown oil (870 mg, 4.3 mmol, 100% yield). 1H NMR (DMSO-d6) δ ppm 5.08 (brs, 2H), 6.75 (d, J=15 Hz, 1H), 6.90-7.01 (m, 2H), 7.07 (dd, J=9 Hz, J=3 Hz, 1H), 7.16 (t, 9 Hz, 1H), 7.26-7.30 (m, 1H), 7.73 (d, J=3 Hz, 1H); ESIMS found for C11H9FN2O m/z 204.9 (M+H).




embedded image


6-(2-Fluorophenoxy)pyridin-3-amine (LXX): Dark brown oil (611 mg, 3.0 mmol, 91% yield). ESIMS found for C11H9FN2O m/z 204.9 (M+H).


Preparation of intermediate 6-phenylpyridin-3-amine (LXXIV) is depicted below in Scheme 16.




embedded image



Step 1


To a solution of 2-bromo-5-nitropyridine (LXXI) (302 mg, 1.49 mmol) in a mixture of dioxane (14 mL) and water (3 mL) was added phenylboronic acid (LXXII) (199 mg, 1.64 mmol), Pd(PPh3)4 (86 mg, 0.74 mmol) and K3PO4 (473 mg, 2.23 mmol). The reaction was microwaved at 95° C. for 1 h. The reaction was cooled and the organic phase was separated, dried over MgSO4 and evaporated under vacuum. The residue was purified by silica gel column chromatography (100% hexane→5:95 EtOAc:hexane) to give 5-nitro-2-phenylpyridine (LXXIII) as off-white needles (254 mg, 1.27 mmol, 85% yield). ESIMS found for C11H8N2O2 m/z 200.9 (M+H).


Step 2


Preparation of intermediate 6-phenylpyridin-3-amine (LXXIV) was performed following the procedure listed in Scheme 11, Step 2. Black green viscous oil (211 mg, 1.24 mmol, 98% yield). 1H NMR (DMSO-d6) δ ppm 5.45 (s, 2H), 6.99 (dd, J=1 Hz, J=3 Hz, 1H), 7.25-7.28 (m, 1H), 7.38-7.40 (m, 2H), 7.62 (d, J=11 Hz, 1H0, 7.89-7.91 (m, 1H), 8.02 (d, J=3 Hz, 1H); ESIMS found for C11H10N2 m/z 171 (M+H).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 16.




embedded image


6-(3-Fluorophenyl)pyridin-3-amine (LXXV): Brown oil (252 mg, 1.34 mmol, 98% yield). ESIMS found for C11H9FN2 m/z 189 (M+H).




embedded image


6-(4-Fluorophenyl)pyridin-3-amine (LXXVI): Deep purple oil (202 mg, 1.07 mmol, 98% yield). ESIMS found for C11H9FN2 m/z 189 (M+H).


Preparation of intermediate 5-benzylpyridin-3-amine (LXXX) is depicted below in Scheme 17.




embedded image



Step 1


To a solution of 3-bromo-5-nitropyridine (LXXVII) (295 mg, 1.45 mmol) in dioxane (14 mL) was added 2-benzyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (LXXVIII) (420 μL, 1.89 mmol), PdCl2(dppf)2, (120 mg, 0.15 mmol) and 2M aqueous K3PO4 (2.2 mL, 4.36 mmol). The reaction was microwaved at 90° C. for 2 h. The reaction was cooled and the organic phase was separated, dried over MgSO4 and evaporated under vacuum. The residue was purified by silica gel column chromatography (100% hexane→6:94 EtOAc:hexane) to give 3-benzyl-5-nitropyridine (LXXIX) as brown oil (117 mg, 0.54 mmol, 37% yield). 1H NMR (DMSO-d6) δ ppm 4.16 (s, 2H), 7.21-7.25 (m, 1H), 7.31-7.33 (m, 4H), 8.45-8.46 (m, 1H), 8.93 (d, J=2 Hz, 1H), 9.21 (d, J=3 Hz, 1H); ESIMS found for C12H10N2O2 m/z 215 (M+H).


Step 2


Preparation of 5-benzylpyridin-3-amine (LXXX) was performed following the procedure listed in Scheme 11, Step 2. Black green viscous oil (139 mg, 0.75 mmol, 98% yield). ESIMS found for C12H12N2 m/z 185 (M+H).


Preparation of intermediate 2-(4-methylpiperazin-1-yl)pyridin-3-amine (LXXXIV) is depicted below in Scheme 18.




embedded image



Step 1


To a microwave vial was added 2-chloro-3-nitropyridine (LXXXI) (1.00 g, 6.31 mmol), 1-methylpiperazine (LXXXII) (0.758 g, 7.57 mmol), cesium carbonate (2.88 g, 8.83 mmol), Pd2(dba)2 (0.173 g, 0.189 mmol), xanthphos (0.109 g, 0.189 mmol), and dioxane (5 mL). The reaction vial was capped and purged with argon. The solution into the reaction vial was heated under microwave irradiation for 2 h at 90° C. The solution was filtered through a pad of Celite and concentrated to a residue under vacuum. The residue was purified by column chromatography (1:99 MeOH:CHCl3→8:92 MeOH:CHCl3) to afford 1-methyl-4-(3-nitro-pyridin-2-yl)-piperazine (LXXXIII) (1.30 g, 5.85 mmol, 93% yield) as a brown oil.


Step 2


To a stirring solution of 1-methyl-4-(3-nitro-pyridin-2-yl)-piperazine (LXXXIII) (1.30 g, 5.85 mmol) in MeOH (15 mL) was added 10% Pd/C. The solution was purged with hydrogen. The solution was stirred at room temperature for 16 h under hydrogen. The solution was filtered through a pad of Celite and concentrated to a residue under vacuum. The residue was purified by column chromatography (100% CHCl3→2:98 MeOH[7N NH3]:CHCl3) to afford 2-(4-methylpiperazin-1-yl)pyridin-3-amine (LXXXIV) (0.466 g, 2.42 mmol, 52% yield) as a tan solid. ESIMS found for C10H16N4 m/z 192.4 (M+H).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 18.




embedded image


6-(Pyrrolidin-1-yl)pyridin-3-amine (LXXXV): Deep purple oil (1.43 g, 8.77 mmol, 100% yield). ESIMS found for C9H13N3 m/z 164 (M+H).




embedded image


6-(4-Methylpiperazin-1-yl)pyridin-3-amine (LXXXVI): Purple solid (598 mg, 3.11 mmol, 32% yield). ESIMS found for C10H16N4 m/z 193 (M+H).




embedded image


6-Morpholinopyridin-3-amine (LXXXVII): Purple solid (782 mg, 4.36 mmol, 95% yield). ESIMS found for C9H13N3O m/z 180 (M+H).




embedded image


N2-(2-(Dimethylamino)ethyl)-N2-methylpyridine-2,5-diamine (LXXXVIII): Deep purple oil (1.55 g, 7.98 mmol, 96% yield). ESIMS found for C10H18N4 m/z 195 (M+H).


Preparation of intermediate 1-(5-aminopyridin-2-ylpiperidin-4-ol (XCI) is depicted below in Scheme 19.




embedded image



Step 1


To a solution of 2-chloro-5-nitropyridine (LXV) (5.0 g, 31.5 mmol) in DMF (50 mL) was added piperidin-4-ol (LXXXIX) (3.5 g, 34.65 mmol) and K2CO3 (8.7 g, 63.0 mmol). The reaction was headed at 85° C. overnight. The solution was poured into ice water, stirred for 15 min and then filtered. The solid was washed with cold water and dried under vacuum to produce 1-(5-aminopyridin-2-ylpiperidin-4-ol (XC) as a yellow solid (6.62 g, 29.67 mmol, 94.2% yield). 1H NMR (DMSO-d6) δ ppm 1.34-1.42 (m, 2H), 1.77-1.83 (m, 2H), 3.40-3.56 (m, 2H), 3.76-3.83 (m, 1H), 4.12 (brd, 2H), 4.81 (d, J=4 Hz, 1H), 6.94 (d, J=10 Hz, 1H), 8.17 (dd, J=10 Hz, J=3 Hz, 1H), 8.94 (d, J=3 Hz, 1H); ESIMS found for C10H13N3O3 m/z 224.1 (M+H).


Step 2


Preparation of intermediate 1-(5-aminopyridin-2-yl)piperidin-4-ol (XCI) was performed following the procedure listed in Scheme 11, Step 2. Dark brown oil (5.7 g, 29.5 mmol, 99.5% yield). 1H NMR (DMSO-d6) δ ppm 1.36 (tq, J=13 Hz, J=4 Hz, 2H), 1.72-1.76 (m, 2H), 2.79 (dt, J=13 Hz, J=3 Hz, 2H), 3.54-3.61 (m, 1H), 3.70-3.78 (m, 2H), 4.49 (s, 2H), 4.61 (d, J=4 Hz, 1H), 6.61 (d, J=9 Hz, 1H), 6.88 (dd, J=9 Hz, J=3 Hz, 1H), 7.57 (d, J=3 Hz, 1H); ESIMS found for C10H15N3O m/z 194.1 (M+H).


The following intermediates were prepared in accordance with the procedure described in the above Scheme 19.




embedded image


6-(Piperidin-1-yl)pyridin-3-amine (XCII): Dark red viscous oil (4.93 g, 27.81 mmol, 95.9% yield). 1H NMR (DMSO-d6) δ ppm 1.48-1.71 (m, 8H), 3.42-3.53 (m, 2H), 4.48 (brs, 2H), 6.59 (d, J=9 Hz, 1H), 6.89 (dd, J=9 Hz. J=3 Hz. 1H), 7.58 (d, J=3 Hz. 1H); ESIMS found for C10H15N3 m/z 178.0 (M+H).




embedded image


5-Methyl-6-(pyrrolidin-1-yl)pyridin-3-amine (XCIII): Dark blue viscous oil (2.06 g, 12.62 mmol, 100% yield). 1H NMR (DMSO-d6) δ ppm 1.76-1.82 (m, 4H), 2.13 (s, 3H), 3.15-3.20 (m, 4H), 4.53 (brs, 2H), 6.74 (d, J=3.5 Hz, 1H), 7.42 (d, J=2.7 Hz, 1H); ESIMS found for C10H15N3 m/z 178.1 (M+H).




embedded image


6-(Azetidin-1-yl)-5-methylpyridin-3-amine (XCIV): Dark red solid (2.0 g, 11.29 mmol, 86.9% yield). 1H NMR (DMSO-d6) δ ppm 2.11 (quin, J=7 Hz, 2H), 3.76-3.87 (m, 4H), 4.50 (brs, 2H), 6.72 (d, J=2.5 Hz, 1H), 7.38 (d, J=2.5 Hz. 1H); ESIMS found for C9H13N3 m/z 164.4 (M+H).




embedded image


6-(Azetidin-1-yl)pyridin-3-amine (XCV): Burgundy solid (1.45 g, 9.70 mmol, 99.3% yield). ESIMS found for C8H11N3 m/z 149.0 (M+H).


Preparation of intermediate tert-butyl 4-(5-aminopyridin-2-ylpiperazine-1-carboxylate (XCVIII) is depicted below in Scheme 20.




embedded image



Step 1


To a solution of 2-chloro-5-nitropyridine (LXV) (2.0 g, 12.6 mmol) in EtOH (20 mL) was added tert-butyl piperazine-1-carboxylate (XCVI) (7.05 g, 37.9 mmol). The reaction was headed at 70° C. for 16 h. The reaction was concentrated under vacuum and then dissolved in EtOAc. The EtOAc was washed with 1 M NaOH, brine and then dried over MgSO4 to give tert-butyl 4-(5-nitropyridin-2-yl)piperazine-1-carboxylate (XCVII) as a yellow solid (4.94 g). ESIMS found for C14H20N4O4 m/z 309.0 (M+H).


Step 2


Preparation of intermediate/m-butyl 4-(5-aminopyridin-2-ylpiperazine-1-carboxylate (XCVIII) was performed following the procedure listed in Scheme 11, Step 2. Purple solid (990 mg, 3.56 mmol, quantitative). ESIMS found for C14H22N4O2 m/z 278.8 (M+H).


Preparation of intermediate N-(3-aminopyridin-4-yl) cyclopropanecarboxamide (CII) is depicted below in Scheme 21.




embedded image



Step 1


Preparation of N-(3-nitropyridin-4-yl)cyclopropanecarboxamide (Cl) was performed following the procedure listed in Scheme 4, Step 1. Orange solid (130 mg, 0.93 mmol, 13% yield). ESIMS found for C9H9N3O3 m/z 207.8 (M+H).


Step 2


Preparation of intermediate N-(3-aminopyridin-4-yl) cyclopropanecarboxamide (CII) was performed following the procedure listed in Scheme 11, Step 2. Dark grey solid (100 mg, 0.56 mmol, quantitative). ESIMS found for C9H11N3O m/z 178.3 (M+H).


Preparation of intermediate (5-aminopyridin-2-ylpyrrolidin-1-ylmethanone (CV) is depicted below in Scheme 22.




embedded image



Step 1


To a solution of 5-nitropicolinic acid (CIII) (500 mg, 2.97 mmol) in DMF (15 mL) was added pyrrolidine (244 μl, 2.47 mmol) and DIPEA (1.03 mL, 5.95 mmol). The reaction was cooled at 0° C. before adding HATU (1.13 g, 2.47 mmol). The reaction was warmed to room temperature and stirred for 2 hrs. The reaction was concentrated under vacuum and then dissolved in a mixture of water and 10% iPrOH/CHCl3. The organic layer was separated and the aqueous phase was washed again with 10% iPrOH/CHCl3. The combined organic phases were washed with brine, dried over MgSO4 and evaporated to yield (5-nitropyridin-2-ylpyrrolidin-1-ylmethanone (CIV) as a red solid (849 mg). ESIMS found for C10H11N3O3 m/z 222.1 (M+H).


Step 2


Preparation of intermediate (5-aminopyridin-2-ylpyrrolidin-1-ylmethanone (CV) was performed following the procedure listed in Scheme 11, Step 2. Yellow solid (708 mg, 7.3 mmol, 96.4% yield). ESIMS found for C10H13N3O m/z 191.4 (M+H).


The following intermediate was prepared in accordance with the procedure described in the above Scheme 22.




embedded image


5-Amino-N-cyclopentylpicolinamide (CVI): Yellow solid (450 mg, 2.19 mmol, 93.7% yield). ESIMS found for C11H15N3O m/z 206.1 (M+H).


Preparation of intermediate 6-(methylsulfonyl)pyridin-3-amine (CIX) is depicted below in Scheme 23.




embedded image



Step 1


To a solution of sodium thiomethoxide in THF (53 mL) and H2O (20 mL) cooled to 0° C. was added 2-chloro-5-nitropyridine (LXV) (5.09 g, 32.09 mmol). The reaction was warmed to room temperature and stirred for 2 hrs. The reaction was poured into ice water and stirred for 10 minutes, filtered, washed with water, dried under vacuum to yield 2-(methylthio)-5-nitropyridine (CVII) as a yellow solid (5.14 g, 30.20 mmol, 94.1%). 1H NMR (DMSO-d6) δ ppm 2.62 (s, 3H), 7.57 (d, J=8.9 Hz, 1H), 8.38 (d, J=8.9 Hz, 1H), 9.22 (d, J=2.7 Hz, 1H); ESIMS found for C6H6N2O2S m/z 171.1 (M+H).


Step 2


To a solution of 2-(methylthio)-5-nitropyridine (CVII) (502 mg, 2.95 mmol) in DCM (60 mL) was mCPBA (1.33 g, 5.90 mmol). The reaction was stirred at room temperature for 1 hr. Two additional portions of mCPBA (2×250 mg) were added at 1 hr intervals for a total reaction time of 4 hr. The reaction was poured into saturated aqueous NaHCO3. The organic phase was separated and washed with water, brine and then dried over MgSO4. The solvent was removed under vacuum to produce crude 2-(methylsulfonyl)-5-nitropyridine (CVIII) (854 mg) which was used without purification for step 3. ESIMS found for C6H6N2O4S m/z 203.0 (M+H).


Step 3


Preparation of intermediate 6-(methylsulfonyl)pyridin-3-amine (CIX) was performed following the procedure listed in Scheme 11, Step 2. The crude product was used as is without purification. ESIMS found for C6H8N2O2S m/z 173.0 (M+H).


Preparation of intermediate 5-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXIV) is depicted below in Scheme 24.




embedded image



Step 1


177-indazole-3-carboxylic acid (CX) (100 g, 617 mmol) in DMF was treated with carbonyldiimidazole (110 g, 678 mmol) at room temperature until the evolution of gas ceased (ca. 15 minutes). The reaction was heated to 60-65° C. for 2 h and then allowed to cool to room temperature. N,O-Dimethylhydroxylamine-HCl (66.2 g, 678 mmol) was added as a solid and the mixture was heated to 65° C. for 3 h. The reaction was concentrated to a paste, taken up in DCM and washed subsequently with water and 2 N HCl. The product could be seen coming out of solution. The solid was filtered and rinsed separately with EtOAc. The EtOAc and DCM layers were separately washed with sodium bicarbonate followed by brine, dried over MgSO4 and concentrated under reduced pressure. The resulting solids were combined, triturated with 1:1 mixture of DCM-ether, filtered, and dried to produce N-methoxy-N-methyl-1H-indazole-3-carboxamide (CXI) as a white solid (100 g, 487 mmol, 79% yield). 1H NMR (DMSO-d6) δ ppm 3.46 (s, 3H), 3.69-3.85 (m, 3H), 7.13-7.31 (m, 1H), 7.41 (t, J=7.25 Hz, 1H), 7.56-7.65 (m, 1H), 7.93-8.08 (m, 1H); ESIMS found for C10H11N3O2 m/z 206 (M+H).


Step 2


To N-methoxy-N-methyl-1H-indazole-3-carboxamide (CXI) (20 g, 97.4 mmol) in DCM (1 L) was added (Bis(trifluoroacetoxy)iodo)benzene (46 g, 107 mmol) followed by portionwise addition of iodine (14.84 g, 58.5 mmol) at room temperature. After 1 h, saturated aqueous NaHSO3 (600 mL) was added and a solid began to precipitate which was filtered and rinsed with excess DCM. The filtrate was washed with brine, dried over MgSO4, concentrated and the remaining solid was triturated with a minimal amount of DCM. The combined solids were dried under vacuum over KOH to produce 5-iodo-N-methoxy-N-methyl-1H-indazole-3-carboxamide (CXII) as a white solid (23.2 g, 70 mmol, 72% yield). 1H NMR (DMSO-d6) δ ppm 3.45 (s, 3H), 3.77 (s, 3H), 7.45-7.54 (m, 1H), 7.66 (dd, J=8.81, 1.51 Hz, 1H), 8.40 (d, J=1.01 Hz, 1H); ESIMS found for C10H10IN3O2 m/z 331 (M+H).


Step 3


A mixture of 5-iodo-N-methoxy-N-methyl-1H-indazole-3-carboxamide (CXII) (16.5 g, 50 mmol), 3,4-dihydro-2H-pyran (10.3 mL, 113 mmol) and PPTS (0.12 g, 0.6 mmol) in DCM was heated to reflux for 5 h. The solution was poured into a saturated aqueous NaHCO3 solution, the layers were separated, and the aqueous layer was extracted with DCM. The combined organic layers were washed with 5% aqueous citric acid and brine, dried over MgSO4, and concentrated. The crude product was purified on a silica gel column (100% EtOAc→3:97 MeOH:DCM) to provide 5-iodo-N-methoxy-N-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXIII) as a white viscous oil (19.1 g, 46 mmol, 92% yield). 1H NMR (DMSO-d6) δ ppm 1.28-1.84 (m, 6H), 3.43 (s, 3H), 3.60-4.04 (s, 5H), 5.86-6.08 (m, 1H), 7.45-7.87 (m, 2H), 8.39 (s, 1H); ESIMS found for C15H18IN3O3 m/z 416 (M+H).


Step 4


Lithium aluminum hydride (160 mg, 4.21 mmol) was added in portions to a cooled (0° C.) solution of 5-iodo-N-methoxy-N-methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXIII) (1.46 g, 3.5 mmol) in THF. Stirring was continued at 0° C. until the reaction was completed, approximately 30 min. The reaction was quenched by the slow addition of EtOAc at 0° C., and the whole mixture was poured into 0.4 N aqueous NaHSO4. The organic layer was washed with brine, dried over MgSO4, concentrated, and purified on a silica gel column (100% EtOAc→3:97 MeOH:DCM) to give 5-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXIV) as a white solid (0.90 g, 3.15 mmol, 72% yield). 1H NMR (DMSO-d6) δ ppm 1.50-1.71 (m, 2H), 1.71-1.87 (m, 1H), 1.97-2.15 (m, 2H), 2.31-2.42 (m, 1H), 3.66-3.99 (m, 2H), 5.96-6.17 (m, 1H), 7.78 (d, J=6 Hz, 1H), 7.84 (d, J=6 Hz, 1H), 8.50 (s, 1H), 10.13 (s, 1H); ESIMS found for C13H13IN2O2 m/z 357 (M+H).


Preparation of intermediate 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXVIII) is depicted below in Scheme 25.




embedded image



Step 1


A suspension of indazole-3-carboxylic acid (CX) (1.0 g, 6.16 mmol) in glacial acetic acid (60 mL) was heated at 120° C. to get a clear solution. The solution was cooled to 90° C. A solution of bromine (0.633 mL, 12.33 mmol) in glacial acetic acid (2 mL) was added slowly to the solution while heating at 90° C. The solution was further heated 16 h at 90° C. The solution was cooled to room temperature, poured into ice water and further stirred at room temperature for 15 min. The solids formed were filtered, washed with cold water and dried under vacuum at room temperature to get 5-bromo-1H-indazole-3-carboxylic acid (CXV) as a white solid (1.30 g, 5.39 mmol, 87.5% yield). 1H NMR (DMSO-d6) δ ppm 13.95 (s, 1H), 13.18 (br s, 1H), 8.21 (d, J=1.2 Hz, 1H), 7.65 (d, J=7.0 Hz, 1H), 7.56 (dd, J=7.0, 1.2 Hz, 1H); ESIMS found for C8H4BrN2O2 m/z 242.0 (M+H).


Step 2


Concentrated sulfuric acid (1 mL) was added to a suspension of 5-bromo-1H-indazole-3-carboxylic acid (CXV) (1.30 g, 5.39 mmol) in dry MeOH (50 mL) and heated to reflux for 4 h under argon. The solution was cooled to room temperature and the MeOH was evaporated under vacuum. The residue was dissolved in EtOAc and washed with water. The organic phase was dried over Na2SO4, filtered and concentrated to afford methyl 5-bromo-1H-indazole-3-carboxylate (CXVI) as a white solid (1.35 g, 5.29 mmol, 98% yield). 1H NMR (DMSO-d6) δ ppm 14.13 (s, 1H), 8.21 (d, J=1.6 Hz, 1H), 7.67 (d, j=7.2 Hz, 1H), 7.59 (dd, j=7.2, 1.2 Hz, 1H), 3.92 (s, 3H); ESIMS found for C9H7BrN2O2 m/z 256.0 (M+H).


Step 3


A suspension of methyl 5-bromo-1H-indazole-3-carboxylate (CXVI) (1.35 g, 5.29 mmol), pyridinium p-toluenesulfonate (0.143 g, 0.56 mmol) and 3,4 dihydro-277-pyran (1.02 mL, 11.90 mmol) in anhydrous dichloroethane (20 mL) was refluxed 5 h under argon. The suspension was turned into the clear solution. The solution was cooled and the excess solvent was evaporated under vacuum. The residue was dissolved in EtOAc and washed with dilute NaHCO3, solution (satd. NaHCO3 soln/H2O: 1:9). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (100% hexanes→5:95 EtOAc:hexanes) to get methyl 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylate (CXVII) as a white solid (1.47 g, 4.34 mmol, 82% yield). 1H NMR (DMSO-d6) δ ppm 8.22 (d, J=1.4 Hz, 1H), 7.89 (d, J=7.2 Hz, 1H), 7.68 (dd, J=7.2, 1.6 Hz, 1H), 6.02 (dd, j=8.0, 2.4 Hz, 1H), 3.94 (s, 3H), 3.88 (m, 1H), 3.79 (m, 1H), 2.37-2.31 (m, 1H), 2.05-1.96 (m, 2H), 1.77-1.73 (m, 1H). 1.60-1.58 (m, 2H); ESIMS found for C14H15BrN2O3 m/z 340.0 (M+H).


Step 4


2 N Aqueous NaOH solution (10 mL) was added to a suspension of methyl 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylate (CXVII) (1.30 g, 3.83 mmol) in water (20 mL) and heated at 90° C. for 1 h. The solution was cooled to room temperature, diluted with ice water and acidified to pH 3.0 with 10% aqueous HCl. The solids formed were filtered, washed with cold water and dried under vacuum at room temperature to get 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXVIII) as a white solid (0.87 g, 2.68 mmol, 70% yield). ESIMS found for C13H13BrN2O3 m/z 326.0 (M+H).


Step 5


To a solution of 5-bromo-1H-indazole-3-carboxylic acid (CXV) (59.8 g, 248 mmol) in THF (800 mL) under argon was added 3,4 dihydro-2H-pyran (50.6 mL, 558 mmol) and p-TsOH (4.72 g, 24.8 mmol). The reaction was heated to reflux at 60° C. for 16 h. An additional portion of p-TsOH (0.025 eq) and 3,4 dihydro-277-pyran (0.56 eq) was added and the reflux continued for 5 h. The solution was concentrated under vacuum. EtOAc was added to the residue and the suspension was filtered and dried under high vacuum overnight to produce 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXVIII) as a white solid (49.07 g, 150.9 mmol, 60.8% yield). ESIMS found for C13H13BrN2O3 m/z 326.3 (M+H).


Preparation of intermediate 5-bromo-1-trityl-1H-indazole-3-carboxylic acid (CXXI) is depicted below in Scheme 26.




embedded image



Step 1


Preparation of intermediate ethyl 5-bromo-1H-indazole-3-carboxylate (CXIX) was performed following the procedure listed in Scheme 25, Step 2. White solid. (3.60 g, 13.38 mmol, 64.5% yield). 1H NMR (DMSO-d6) δ ppm 1.37 (t, J=7 Hz, 3H), 4.40 (q, J=7 Hz. 2H), 7.57 (dd, J=9 Hz, J=2 Hz. 1H), 7.66 (d, J=9 Hz, 1H), 8.20 (d, J=2 Hz. 1H), 14.11 (brs, 1H); ESIMS found for C10H9BrN2O2 m/z 269.0 (M+H).


Step 2


To a solution of ethyl 5-bromo-1H-indazole-3-carboxylate (CXIX) and trityl chloride in DCM was slowly added DIPEA. The solution was stirred at room temperature overnight. The reaction was poured into water and stirred for 5 min. The organic layer was separated, dried over MgSO4 and concentrated under vacuum. The residue was purified by column chromatography using a ISCO 200RF system with a SiO2 column (12 g) (100% hexanes→10:90 EtOAc:hexanes) to produce a white solid. (357 mg, 0.70 mmol, 69.8% yield). 1H NMR (DMSO-de) δ ppm 1.34 (t, J=7 Hz, 3H), 4.38 (q, J=7 Hz, 2H), 6.43 (d, J=9.5 Hz, 1H), 7.11-7.14 (m, 6H), 7.31-7.35 (m, 10H), 8.23 (d, J=2 Hz, 1H); ESIMS found for C29H23BrN2O2 m/z 511.0 (M+H).


Step 3


Preparation of intermediate 5-bromo-1-trityl-1H-indazole-3-carboxylic acid (CXXI) by hydrolysis of ethyl 5-bromo-1-trityl-1H-indazole-3-carboxylate (CXX) can be performed following the procedure listed in Scheme 25, Step 3.


Step 4


Preparation of intermediate 5-bromo-1-trityl-1H-indazole-3-carboxylic acid (CXXI) by tritylation of 5-bromo-1H-indazole-3-carboxylic acid (CXV) can be performed following the procedure listed in the Journal of Medicinal Chemistry (2003), 46(25), 5458-5470.


Example 1

Preparation of 5-(5-(3,3-dimethylureido)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (1) is depicted below in Scheme 27.




embedded image



Step 1-2


A solution of 5-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXIV) (1.780 g, 5.0 mmol), bis(pinacolato)diboron (1.523 g, 6.0 mmol), KOAc (1.471 g, 15 mmol) and dry DMF (20 mL) was purged with argon. PdCl2(dppf)2 (0.245 g, 0.3 mmol) was added to the reaction and purged again with argon. The solution was heated at 90° C. for 2 h. Once TLC showed the disappearance of (CXIV), the solution was cooled to room temperature. To this solution was added K3PO4 (1.592 g, 7.5 mmol), 3-(5-bromopyridin-3-yl)-1,1-dimethylurea (XXII) (1.220 g, 5.0 mmol), Pd(PPh3)4 (173 mg, 0.15 mmol) and water (2 mL). The solution was purged with argon and heated at 90° C. for 3 h. The solution was cooled to room temperature and then concentrated under reduced pressure. The residue was dissolved in DCM and washed with water, dried over MgSO4, filtered and then evaporated under vacuum. The residue was purified on a silica gel column (100% DCM→2:98 MeOH:DCM) to give 3-(5-(3-formyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)pyridin-3-yl)-1,1-dimethylurea (CXXIII) as a brown viscous oil which solidified under vacuum at room temperature (354 mg, 0.90 mmol, 18% yield for 2 steps). 1H NMR (DMSO-d6) δ ppm 10.22 (s, 1H), 8.76 (d, J=1.6 Hz, 1H), 8.63 (s, 1H), 8.52 (d, J=1.6 Hz, 1H), 8.36 (s, 1H), 8.24 (m, 1H), 8.05 (d, J=7.2 Hz, 1H), 7.91 (dd, J=7.2, 1.4 Hz, 1H), 6.13 (dd, J=7.6, 2.0 Hz, 1H), 3.93 (m, 1H), 3.85 (m, 1H), 2.98 (s, 6H), 2.47-2.42 (m, 1H), 2.11-2.06 (m, 2H), 1.82-1.79 (m, 1H) 1.64 (m, 2H); ESIMS found for C21H23N5O3 m/z 394.0 (M+H).


Step 3


A solution of sodium hydroxide (0.173 g, 4.33 mmol) in water (5 mL) was added to a solution of silver nitrate (0.367 g, 2.16 mmol) in water (5 mL) to give a brown precipitate. 3-(5-(3-formyl-1-(tetrahydro-2H-pyran-2-yl)-1H-indazol-5-yl)pyridin-3-yl)-1,1-dimethylurea (CXXIII) (0.340 g, 0.86 mmol) was dissolved in 1,4-dioxane (10 mL) and added to the reaction which was stirred overnight at room temperature. The solution was diluted with water and then extracted with diethyl ether. The aqueous layer was separated and carefully brought to pH=3 with aqueous HCl. The aqueous layer was then extracted with 10% iPrOH/chloroform. The combined organic layers were then dried (Na2SO4), filtered and concentrated to give 5-(5-(3,3-dimethylureido)pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXXIV) as a brownish white solid (246 mg, 0.60 mmol, 70% yield). 1H NMR (DMSO-d6) δ ppm 13.26 (hr. s, 1H), 8.87 (s, 1H), 8.63 (s, 1H), 8.41 (s, 1H), 8.34 (s, 1H), 8.03 (d, J=7.1 Hz, 1H), 7.86 (dd, J=7.2, 1.3 Hz, 1H), 6.06 (dd, J=8.0, 4.0 Hz, 1H), 3.92 (m, 1H), 3.80 (m, 1H), 2.98 (s, 6H), 2.42-2.39 (m, 1H), 2.03-2.02 (m, 2H), 1.79-1.77 (m, 1H) 1.61 (m, 2H); ESIMS found for C21H23N5O4 m/z 410.0 (M+H).


Step 4


HATU (0.190 g, 0.5 mmol) was added to a solution of 5-(5-(3,3-dimethylureido)pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXXIV) (0.39 g, 1.21 mmol) and diisopropylethylamine (0.174 mL, 1.0 mmol) in DMF stirred at room temperature under argon. After stirring 5 min, the solution was added with 3-aminopyridine (CXXV) (0.047 g, 0.5 mmol). The solution was stirred overnight at room temperature under argon. The DMF was removed under reduced pressure, and the residue was treated with water, sonicated briefly and filtered. The solids were washed with cold water and dried at room temperature. The product was purified by column chromatography using a 4 g Thomson normal phase silica gel cartridge (100% DCM→5:95 MeOH:DCM) to afford 5-(5-(3,3-dimethylureido)pyridin-3-yl)-N-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXXVI) as an off white solid (323 mg, 0.67 mmol, 55% yield). 1H NMR (DMSO-d6) δ ppm 10.56 (s, 1H), 9.06 (d, J=2.0 Hz, 1H), 8.75 (d, J=1.6 Hz, 1H), 8.64 (s, 1H), 8.53 (d, J=1.6 Hz, 1H), 8.46 (s, 1H), 8.34-8.29 (m, 2H), 8.26 (m, 1H), 8.03 (d, J=7.0 Hz, 1H), 7.88 (dd, J=7.0, 1.2 Hz, 1H), 7.43 (dd, J=6.64, 3.84 Hz, 1H), 6.07 (dd, J=8.0, 1.8 Hz, 1H), 3.98 (m, 1H), 3.82 (m, 1H), 2.98 (s, 6H), 2.63-2.60 (m, 1H), 2.11-2.06 (m, 2H), 1.83-1.81 (m, 1H) 1.52 (m, 2H); ESIMS found for C21H23N5O4 m/z 410.0 (M+H).


Step 5


TFA (5 mL) was added to a solution of 5-(5-(3,3-dimethylureido)pyridin-3-yl)-N-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXXVI) (0.134 g, 0.27 mmol) and triethylsilane (0.110 mL, 0.69 mmol) in DCM (5 mL) and stirred 3 h at room temperature. The solvent was removed under vacuum. The residue was treated with water, sonicated briefly to disperse the solids, basified to pH 9.0 with 5 N NH4OH and sonicated again. The solids were filtered, washed with cold water and purified by column chromatography (100% DCM→5:95 MeOH[7N NH3]:DCM) to afford 5-(5-(3,3-dimethylureido)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (1) as a white solid (35.8 mg, 0.09 mmol, 33% yield). 1H NMR (DMSO-d6) δ ppm 13.99 (s, 1H), 10.69 (s, 1H), 9.08 (d, J=1.2 Hz, 1H), 8.74 (d, J=1.8 Hz, 1H), 8.63 (s, 1H), 8.51 (d, J=1.5 Hz, 1H), 8.47 (s, 1H), 8.33-8.30 (m, 2H), 8.26 (m, 1H), 7.80 (s, 2H), 7.41 (dd, J=6.6, 3.6 Hz, 1H), 2.98 (s, 6H); ESIMS found for C21H19N7O2 m/z 402.3 (M+H).


The following compound was prepared in accordance with the procedure described in the above Example 1.




embedded image


N-(5-Fluoropyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 23.


Light tan solid. 1H NMR (DMSO-d6) δ ppm 7.53 (dd, J=8 Hz, J=5 Hz, 1H), 7.82-7.86 (m, 2H), 8.13-8.15 (m, 1H), 8.31-8.34 (m, 2H), 8.47-8.48 (m, 1H), 8.60 (dd, J=5 Hz, J=2 Hz, 1H), 894 (d, J=2 Hz, 1H), 8.99 (d, J=2 Hz, 1H), 10.97 (s, 1H), 14.05 (s, 1H); ESIMS found for C18H12FN5O m/z 334 (M+1).


Example 2

Preparation of 5-(5-fluoropyridin-3-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (2) is depicted below in Scheme 28.




embedded image



Step 1


HATU (1.125 g, 2.96 mmol) was added to a solution of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXVIII) (0.876 g, 2.69 mmol) and diisopropylethylamine (1.03 mL, 5.92 mmol) in DMF stirred at room temperature under argon. After stirring 5 min, the solution was added with 5-amino-2-trifluoromethyl pyridine (CXXVII) (0.479 g, 2.96 mmol). The solution was stirred 24 h at room temperature under argon. The DMF was removed under reduced pressure, and the residue was treated with water, sonicated briefly and filtered. The solids were washed with cold water and dried at room temperature. The product was purified by silica gel column chromatography (100% hexanes→7:93 EtOAc:hexanes) to afford 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (CXXVIII) as a white solid (1.17 g, 2.50 mmol, 93% yield). 1H NMR (DMSO-d6) 5 ppm 10.93 (s, 1H), 9.23 (d, J=1.9 Hz, 1H), 8.60 (dd, J=6.8, 1.4 Hz, 1H), 8.38 (d, J=4.4 Hz, 1H), 7.95 (m, 2H), 7.70 (dd, J=7.1, 1.5 Hz, 1H), 6.04 (dd, J=8.1, 1.9 Hz, 1H), 3.98 (m, 1H), 3.82 (m, 1H), 2.59-2.54 (m, 1H), 2.08-2.03 (m, 2H), 1.81-1.77 (m, 1H). 1.66-1.61 (m, 2H); ESIMS found for C19H16BrF3N4O2 m/z 470.0 (M+H).


Step 2


A solution of 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-N-(6-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide (CXXVIII) (0.469 g, 1 mmol), 5-fluoro-pyridyl-3-boronic acid (CXXIX) (0.156 g, 1.1 mmol), potassium phosphate tribasic (0.318 g, 1.5 mmol) and water (degassed, 1 mL) in DMF (10 mL) was purged with argon. Tetrakis(triphenylphosphine)palladium(0) (0.034 g, 0.03 mmol) was added and the solution was purged again with argon. The reaction was heated to 90° C. for 3 h when TEC showed disappearance of starting material. The solution was cooled to room temperature and excess solvent was removed under vacuum. The residue was treated with water, sonicated briefly and the solids formed were filtered. The solids were washed with cold water and dried under vacuum at room temperature which was purified by silica gel column chromatography (2:8 EtOAc:hexanes→3:7 EtOAc:hexanes) to afford 5-(5-fluoropyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-N-(6-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide (CXXX) as a white solid (427 mg, 0.88 mmol, 88% yield). 1H NMR (DMSO-d6) δ ppm 10.95 (s, 1H), 9.25 (d, J=1.8 Hz, 1H), 8.85 (m, 1H), 8.63 (d, J=1.8 Hz, 1H), 8.61 (d, J=2.1 Hz, 1H), 8.53 (m, 1H), 8.16-8.13 (m, 1H), 8.08 (d, J=7.1 Hz, 1H), 7.97-7.94 (m, 2H), 6.11 (dd, J=8.1, 1.8 Hz, 1H), 4.01 (m, 1H), 3.88-3.83 (m, 1H), 2.63-2.60 (m, 1H), 2.11-2.07 (m, 2H), 1.83-1.80 (m, 1H). 1.69-1.65 (m, 2H); ESIMS found for C24H19F4N5O2 m/z 486.0 (M+H).


Step 3


TFA (10 mL) was added to a solution of 5-(5-fluoropyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (CXXX) (0.420 g, 0.86 mmol) and triethylsilane (0.345 mL, 2.16 mmol) in DCM (10 mL) and stirred 5 h at room temperature. The solvent was removed under vacuum. The residue was treated with water, sonicated briefly to disperse the solids, basified to pH 9.0 with 5 N NH4OH and sonicated again. The solids were filtered, washed with cold water and air dried at room temperature. The solids were suspended in DCM:MeOH (1:1) mixture and boiled to get a clear solution. The solution was cooled to room temperature. The solids formed were filtered washed with DCM and dried under vacuum at room temperature to get 5-(5-fluoropyridin-3-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (2) as a white solid (72.9 mg, 0.18 mmol, 21% yield). 1H NMR (DMSO-d6) δ ppm 14.13 (hr. s, 1H), 11.11 (s, 1H), 9.27 (d, J=1.8 Hz, 1H), 8.84 (m, 1H), 8.63 (dd, J=6.8, 1.8 Hz, 1H), 8.60 (d, J=2.0 Hz, 1H), 8.53 (m, 1H), 8.14-8.11 (m, 1H), 7.94 (d, J=6.9 Hz, 1H), 7.90-7.83 (m, 2H); ESIMS found for C19H11F4N5O m/z 402.30 (M+H).


The following compound was prepared in accordance with the procedure described in the above Example 2.




embedded image


5-(Pyridin-3-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 3.


White solid (19% yield). 1H NMR (DMSO-d6) δ ppm 14.03 (hr. s, 1H), 11.10 (s, 1H), 9.27 (d, J=1.8 Hz, 1H), 8.94 (d, J=1.6 Hz, 1H), 8.63 (dd, J=6.8, 1.7 Hz, 1H), 8.60 (m, 1H), 8.48 (s, 1H), 8.15-8.13 (m, 1H), 7.93 (d, J=6.9 Hz, 1H), 7.85 (s, 2H), 7.54 (m, 1H); ESIMS found for C19H12F3N5O m/z 384.0 (M+H).




embedded image


N-(1-Methyl-5-(trifluoromethyl)-1H-pyrazol-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 37.


Light green solid (76.7 mg, 0.20 mmol, 48.4% yield). NMR (DMSO-d6) 5 ppm 3.93 (s, 3H), 7.18 (s, 1H), 7.55 (dt, J=8 Hz, J=3 Hz, 1H), 7.81 (dd, J=15 Hz, J=9 Hz, 2H), 8.16 (d, J=8 Hz, 1H), 8.45 (s, 1H), 8.61 (d, J=4 Hz, 1H), 8.95 (s, 1H), 10.81 (s, 1H), 13.96 (s, 1H); ESIMS found for C18H13F3N6O m/z 387.1 (M+H).




embedded image


5-(Pyridin-3-yl)-N-(pyridin-3-ylmethyl)-1H-indazole-3-carboxamide 42.


White solid (54.5 mg, 0.17 mmol, 78% yield). 1H NMR (DMSO-d6) δ ppm 4.53 (d, J=6 Hz, 2H), 7.35 (dd, J=8 Hz, J=5 Hz, 1H), 7.49-7.52 (m, 1H), 7.74-7.78 (m, 3H), 8.09-8.11 (m, 1H), 8.41-8.42 (m, 1H), 8.45 (dd, J=5 Hz, J=2 Hz, 1H), 8.57 (dd, J=5 Hz, J=2 Hz, 1H), 8.59 (d, J=2 Hz, 1H), 8.90 (d, J=2 Hz. 1H), 9.16 (t, J=6 Hz. 1H), 13.77 (s, 1H); ESIMS found for C19H15N5O m/z 330 (M+H).




embedded image


5-(Pyridin-3-yl)-N-(4-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 48.


White solid (67 mg, 0.17 mmol, 62% yield). 1H NMR (DMSO-d6) δ ppm 7.52 (dd, J=8 Hz, J=5 Hz, 1H), 7.83-7.87 (m, 3H), 8.12 (td, J=8 Hz, J=2 Hz, 1H), 8.41 (t, J=1 Hz, 1H), 8.59 (dd, J=5 Hz, J=2 Hz, 1H), 8.75 (d, J=5 Hz, 1H), 8.92 (d, J=3 Hz, 1H), 9.08 (s, 1H), 10.21 (s, 1H), 14.06 (brs, 1H); ESIMS found for C19H12F3N5O m/z 384.0 (M+H).




embedded image


5-(Pyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 53.


Beige solid (23.8 mg, 0.06 mmol, 44.5% yield). 1H NMR (DMSO-d6) δ ppm 1.92-1.97 (m, 4H), 3.38 (t, J=7 Hz, 4H), 6.46 (d, J=9 Hz, 1H), 7.52 (dd, J=8 Hz, J=5 Hz, 1H), 7.80 (dq, J=9 Hz, J=2 Hz, 2H), 7.97 (dd. J=9 Hz. J=3 Hz. 1H), 8.12 (dd. J=8 Hz, J=4 Hz, 1H), 8.47 (s, 1H), 8.50 (d, J=3 Hz, 1H), 8.59 (dd, J=5 Hz, J=2 Hz, 1H), 8.92 (d, J=2 Hz, 1H), 10.22 (s, 1H), 13.86 (s, 1H); ESIMS found for C22H20N6O m/z 385.1 (M+H).




embedded image


N-(4-(Cyclopropanecarboxamido)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 58.


White solid (32.7 mg, 0.08 mmol, 37.0% yield). 1H NMR (DMSO-d6) δ ppm 0.84-0.88 (m, 2H), 0.88-0.92 (m, 2H), 1.91-1.99 (m, 1H), 7.52 (dd, J=8 Hz, J=5 Hz, 1H), 7.73 (d, J=6 Hz, 1H), 7.82 (dd, J=12 Hz. J=9 Hz. 2H), 8.12 (dt, J=9 Hz, J=4 Hz. 1H), 8.34 (d, J=6 Hz, 1H), 8.44 (s, 1H), 8.59 (dd, J=5 Hz, J=2 Hz, 1H), 8.80 (s, 1H), 8.92 (d, J=2 Hz, 1H), 10.03 (s, 1H), 10.31 (s, 1H), 13.98 (s, 1H); ESIMS found for C22H18N6O2 m/z 399.0 (M+H).




embedded image


N-(6-(Cyclopentylcarbamoyl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 181.


Light yellow solid (18 mg, 0.04 mmol, 16.6% yield). NMR (DMSO-d6) 5 ppm 1.50-1.64 (m, 4H), 1.67-1.76 (m, 2H), 1.85-1.94 (m, 4H), 4.24 (quin, J=8 Hz, 1H), 7.53 (dd, J=8 Hz, J=5 Hz, 1H), 7.84 (ABq, 2H), 8.03 (d, J=9 Hz, 1H), 8.14 (d, J=8 Hz, 1H), 8.45 (d, J=8 Hz, 1H), 8.48 (s, 1H), 8.54 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.60 (d, J=4 Hz, 1H), 8.94 (d, J=2 Hz, 1H), 9.16 (d, J=2 Hz, 1H), 10.97 (s, 1H), 14.08 (brs, 1H); ESIMS found for C24H22N6O2 m/z 427.1 (M+H).


Example 3

Preparation of N,5-di(pyridin-3-yl)-1H-indazole-3-carboxamide (4) is depicted below in Scheme 29.




embedded image



Step 1


5-Iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXIV) (1.53 g, 4.30 mmol), pyridine-3-boronic acid (CXXXI) (0.58 g, 4.73 mmol), and potassium phosphate tribasic (1.37 g, 6.45 mmol) was dissolved in 1,4-dioxane (43.0 mL) and water (9.0 mL). Tetrakis(triphenylphosphine)palladium(0) (0.50 g, 0.4301 mmol) was added, and the reaction was heated to 95° C. for 2.5 h. The solvent was removed, and the residue was partitioned between EtOAc and water. The organic phase was separated and washed sequentially with water and brine. The material was dried (MgSO4), concentrated, and purified by flash chromatography using a 40 g Thomson normal phase silica gel cartridge (100% hexanes→1:1 EtOAc:hexanes) to afford 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXXXII) (0.62 g, 2.02 mmol, 47% yield) as a tan amorphous solid. 1H NMR (DMSO-d6) δ ppm 10.23 (s, 1H), 8.95 (d, J=2.3 Hz, 1H), 8.61 (dd, j=4.8, 1.5 Hz, 1H), 8.39 (d, J=0.98 Hz, 1H), 8.17-8.14 (m, 1H), 8.06 (d, J=8.8 Hz, 1H), 7.95-7.93 (m, 1H), 7.64-7.60 (m, 1H), 6.13 (dd, J=9.4, 2.4 Hz, 1H), 3.93-3.90 (m, 1H), 3.86-3.81 (m, 1H), 2.45-2.41 (m, 1H), 2.11-2.07 (m, 2H), 1.82-1.78 (m, 1H), 1.66-1.62 (m, 2H); ESIMS found for C18H17N3O2 m/z 308 (M+H).


Step 2


To a solution of silver nitrate (0.55 g, 3.25 mmol) in water (10 mL) was added a solution of sodium hydroxide (0.26 g, 6.50 mmol) in water (5 mL) to give a brown precipitate. 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carbaldehyde (CXXXII) (0.40 g, 1.30 mmol) dissolved in 1,4-dioxane (3 mL) was added to the reaction which was stirred at room temperature for 2 h. The reaction was then extracted with diethyl ether. The aqueous layer was separated and carefully brought to pH=3 with 10% aqueous HCl. The aqueous layer was then extracted five times with iPrOH/chloroform (1/9). The combined organic layers were then dried (MgSO4) and concentrated to afford 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXXXIII) (0.30 g, 0.93 mmol, 70% yield) as a white solid. 1H NMR (DMSO-d6) δ ppm 13.28 (br, 1H), 8.93 (s, 1H), 8.60 (d, J=4.1 Hz, 1H), 8.32 (d, J=0.83 Hz, 1H), 8.14-8.12 (m, 1H), 8.00 (d, J=8.8 Hz, 1H), 7.86 (dd, j=8.8, 1.7 Hz, 1H), 7.52 (dd, j=7.8, 4.7 Hz, 1H), 6.04 (dd, j=9.3, 2.3 Hz, 1H), 3.92-3.90 (m, 1H), 3.83-3.78 (m, 1H), 2.44-2.37 (m, 1H), 2.08-2.02 (m, 2H), 1.79-1.76 (m, 1H), 1.63-1.61 (m, 2H).


Step 3


To a solution of 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXXXIII) (0.39 g, 1.21 mmol) and 3-aminopyridine (CXXVII) (0.11 g, 1.21 mmol) in DMF (4.0 mL) was added N,N-diisopropylethylamine (0.42 mL, 1.21 mmol). The solution was cooled to 0° C. before adding HATU (0.46 g, 1.21 mmol). The ice bath was removed, and the reaction warmed to room temperature and stirred for 2 h. The DMF was removed under reduced pressure, and the residue was partitioned between chloroform and water. The organic phase was separated and washed sequentially with water and brine, dried over MgSO4, filtered, and concentrated. The product was purified by column chromatography using a 25 g Thomson normal phase silica gel cartridge (100% CHCl3→2:98 MeOH: CHCl3) to afford N,5-di(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXXXIV) (0.36 g, 0.90 mmol, 75% yield) as an off-white solid. 1H NMR (DMSO-d6) δ ppm 10.56 (s, 1H), 9.06 (d, J=2.4 Hz, 1H), 8.94 (d, J=2.3 Hz, 1H), 8.60 (dd, j=4.8, 1.5 Hz, 1H), 8.47 (d, J=1.1 Hz, 1H), 8.34-8.33 (m, 1H), 8.31-8.29 (m, 1H), 8.16-8.14 (m, 1H), 8.04 (d, J=8.8 Hz, 1H), 7.90 (dd, J=8.8, 1.8 Hz, 1H), 7.54-7.52 (m, 1H), 7.43-7.41 (m, 1H), 7.43-7.41 (m, 1H), 6.08-6.06 (m, 1H), 4.01-3.99 (m, 1H), 3.87-3.82 (m, 1H), 2.64-2.57 (m, 1H), 2.11-2.06 (m, 2H), 1.84-1.80 (m, 1H), 1.69-1.65 (m, 2H); ESIMS found for C23H21N5O2 m/z 400 (M+H).


Step 4


TFA (5.0 mL) was added to a solution of N,5-di(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXXXIV) (0.36 g, 0.90 mmol) and triethylsilane (0.29 mL, 1.81 mmol) in DCM (5.0 mL). The solution was stirred overnight at room temperature. An additional 5.0 mL of TFA was added, and the solution was again stirred overnight. The solvents were removed, and the residue was treated with 7 N ammonia in MeOH. The solvents were again removed, and the product was purified by flash chromatography using a 12 g Thomson normal phase silica gel cartridge (100% CHCl3→5:95 MeOH[7N NH3]:CHCl3) to afford N,5-di(pyridin-3-yl)-1H-indazole-3-carboxamide (4) (0.23 g, 0.73 mmol, 82% yield) as a white solid. 1H NMR (DMSO-d6) δ ppm 14.00 (s, 1H), 10.69 (s, 1H), 9.08 (d, J=2.0 Hz, 1H), 8.93 (d, J=2.0 Hz, 1H), 8.60 (dd, J=4.8, 1.3 Hz, 1H), 8.48-8.47 (m, 1H), 8.33-8.31 (m, 2H), 8.15-8.12 (m, 1H), 7.85-7.81 (m, 2H), 7.54-7.51 (m, 1H), 7.41-7.39 (m, 1H); ESIMS found for C18H13N5O m/z 316 (M+H).


The following compounds were prepared in accordance with the procedure described in the above Example 3.




embedded image


N-(3′-Fluorobiphenyl-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 5.


White solid (77 mg, 0.19 mmol, 69% yield). 1H NMR (DMSO-d6) δ ppm 13.95 (s, 1H), 10.50 (s, 1H), 8.94 (d, J=2.3 Hz, 1H), 8.59 (dd, J=4.6, 1.5 Hz, 1H), 8.51 (d, J=1.0 Hz, 1H), 8.31-8.30 (m, 1H), 8.15-8.13 (m, 1H), 7.99-7.97 (m, 1H), 7.83-7.82 (m, 2H), 7.55-7.45 (m, 6H), 7.24-7.22 (m, 1H); ESIMS found for C25H17FN4O m/z 409 (M+H).




embedded image


5-(Pyridin-3-yl)-N-(pyridin-4-yl)-1H-indazole-3-carboxamide 6.


Off-white solid (52 mg, 0.16 mmol, 77% yield). 1H NMR (DMSO-d6) δ ppm 14.05 (br, 1H), 10.83 (s, 1H), 8.94 (d, J=2.0 Hz, 1H), 8.60 (dd, J=4.6, 1.2 Hz, 1H), 8.48-8.47 (m, 3H), 8.15-8.13 (m, 1H), 7.94 (dd, J=5.0, 1.4 Hz, 2H), 7.86-7.82 (m, 2H), 7.54-7.52 (m, 1H); ESIMS found for C18H13N5O m/z 316 (M+H).




embedded image


N-(5-((Dimethylamino)methyl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 7.


Off-white solid (37 mg, 0.10 mmol, 47% yield). 1H NMR (DMSO-d6) δ ppm 14.00 (s, 1H), 10.68 (s, 1H), 8.94 (d, J=2.0 Hz, 1H), 8.91 (d, J=2.3 Hz, 1H), 8.60 (dd, J=4.7, 1.2 Hz, 1H), 8.49-8.48 (m, 1H), 8.38-8.37 (m, 1H), 8.21 (d, J=2.2 Hz, 1H), 8.16-8.13 (m, 1H), 7.85-7.81 (m, 2H), 7.52 (dd, J=7.9, 4.9 Hz, 1H), 3.44 (s, 2H), 2.19 (s, 6H); ESIMS found for C21H20N6O m/z 373 (M+H).




embedded image


5-(Pyridin-3-yl)-N-(6-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 8.


Off-white solid (38 mg, 0.10 mmol, 77% yield). 1H NMR (DMSO-d6) δ ppm 13.99 (hr, 1H), 10.64 (s, 1H), 8.96 (d, J=2.5 Hz, 1H), 8.93 (d, J=2.4 Hz, 1H), 8.59 (dd, J=4.8, 1.5 Hz, 1H), 8.48 (d, J=1.2 Hz, 1H), 8.27 (dd, J=8.5, 2.5 Hz, 1H), 8.16-8.12 (m, 1H), 7.84-7.80 (m, 2H), 7.54-7.51 (m, 1H), 7.41 (d, J=8.5 Hz, 1H), 2.37 (s, 2H), 2.50-2.47 (m, 4H), 1.72-1.70 (m, 4H); ESIMS found for C23H22N6O m/z 399 (M+H).




embedded image


N-(5-(3-Fluorophenyl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 9.


White solid (35 mg, 0.09 mmol, 47% yield). 1H NMR (DMSO-d6) δ ppm 14.05 (hr s, 1H), 10.79 (s, 1H), 9.13 (d, J=2.0 Hz, 1H), 8.94 (d, J=1.9 Hz, 1H), 8.68-8.65 (m, 2H), 8.60 (dd, J=4.83, 4.83 Hz, 1H), 8.52-8.49 (m, 1H), 8.16-8.12 (m, 1H), 7.85-7.81 (m, 2H), 7.62-7.56 (m, 3H), 7.54-7.50 (m, 1H), 7.31-7.26 (m, 1H). ESIMS found for C24H16FN5O m/z 410.5 (M+H).




embedded image


N-(2-(4-Methylpiperazin-1-yl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 11.


White solid (11 mg, 0.03 mmol, 65% yield). 1H NMR (DMSO-d6) δ ppm 14.10 (s, 1H), 9.63 (s, 1H), 8.93 (d, J=2.1 Hz, 1H), 8.65-8.59 (m, 2H), 8.48 (s, 1H), 8.16-8.12 (m, 1H), 8.11-8.09 (m, 1H), 7.87-7.80 (m, 2H), 7.55-7.51 (m, 1H), 7.20-7.17 (m, 1H), 3.10-3.06 (m, 4H), 2.80-2.40 (m, 4H), 2.30 (s, 3H). ESIMS found for C23H23N7O m/z 414.0 (M+H).




embedded image


N-(6-(4-Methylpiperazin-1-yl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 12.


White solid (31 mg, 0.07 mmol, 39% yield). 1H NMR (DMSO-d6) δ ppm 13.86 (br s, 1H), 10.33 (s, 1H), 8.92 (d, J=2.1 Hz, 1H), 8.60-8.58 (m, 2H), 8.46 (s, 1H), 8.14-8.11 (m, 1H), 8.10-8.02 (m, 1H), 7.83-7.78 (m, 2H), 7.54-7.50 (m, 1H), 6.86 (d, J=9.1 Hz, 1H), 3.45-3.42 (m, 4H), 2.42-2.39 (m, 4H), 2.21 (s, 3H). ESIMS found for C23H23N7O m/z 414.3 (M+H).




embedded image


N-(Pyridazin-4-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 14.


Off-white solid (50 mg, 0.16 mmol, 99% yield), 1H NMR (DMSO-d6) δ ppm 14.20-13.90 (br, 1H), 11.15 (s, 1H), 9.71-9.70 (m, 1H), 9.09-9.08 (m, 1H), 8.94 (d, J=2.0 Hz, 1H), 8.61-8.60 (m, 1H), 8.47-8.46 (m, 1H), 8.25 (dd, J=5.9, 2.8 Hz, 1H), 8.16-8.13 (m, 1H), 7.86-7.85 (m, 2H), 7.53 (dd, J=7.8, 5.0 Hz, 1H); ESIMS found for C17H12N6O m/z 317 (M+H).




embedded image


N-(6-((4-Methylpiperazin-1-yl)methyl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 15.


White solid (42 mg, 0.10 mmol, 81% yield). 1H NMR (DMSO-d6) δ ppm 13.97 (br, 1H), 10.65 (s, 1H), 8.97 (d, J=2.4 Hz, 1H), 8.93 (d, J=2.1 Hz, 1H), 8.59 (dd, J=4.7, 1.5 Hz, 1H), 8.48-8.47 (m, 1H), 8.28 (dd, J=8.5, 2.5 Hz, 1H), 8.15-8.12 (m, 1H), 7.85-7.81 (m, 2H), 7.54-7.51 (m, 1H), 7.40 (d, J=8.5 Hz, 1H), 3.55 (s, 2H), 2.42-2.28 (m, 8H), 2.15 (s, 3); ESIMS found for C24H25N7O m/z 428 (M+H).


Example 4

Preparation of 5-(5-fluoropyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (13) is depicted below in Scheme 30.




embedded image


To a stirring solution of 3-aminopyridine (CXXV) (0.195 g, 0.2.07 mmol) in DMF (10 mL) was added 5-bromo-177-indazole-3-carboxylic acid (CXV) (0.500 g, 0.2.07 mmol) and N,N-diisopropylethylamine (0.723 mL, 4.15 mmol). The reaction mixture was cooled to 0° C. and added with HATU (0.787 g, 2.07 mmol). The reaction mixture was allowed to warm to room temperature and stirred for an additional 2 h. The solution was concentrated under vacuum. The residue was purified by column chromatography (1:99 MeOH[7N NH3]:CHCl3→4:96 MeOH[7N NH3]:CHCl3) to afford 5-bromo-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (CXXXV) (0.200 g, 0.63 mmol, 30% yield) as a white solid. ESIMS found for C13H9BrN4O m/z 318.0 (M+H).


Step 2


To a microwave vial was added 5-bromo-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (CXXXV) (0.200 g, 0.63 mmol), 5-fluoropyridine-3-boronic acid (CXXIX) (0.098 g, 0.694 mmol), tetrakistriphenylphosphine)palladium(0) (0.036 g, 0.032 mmol), potassium phosphate (0.201 g, 0.947 mmol), water (1 mL), and DMF (5 mL). The reaction vial was capped, purged with argon and heated under microwave irradiation for 1 h at 180° C. The solution was filtered through a pad of Celite and concentrated under vacuum. The crude product was purified by column chromatography (100% CHCl3→2:98 MeOH[7N NH3]: CHCl3) to afford 5-(5-fluoropyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide (13) (4 mg, 0.01 mmol, 2% yield) as a white solid. 1H NMR (DMSO-d6) δ ppm 14.02 (hr s, 1H), 10.70 (s, 1H), 9.08 (d, J=2.5 Hz, 1H), 8.83 (t, J=1.8 Hz, 1H), 8.60 (d, J=2.7 Hz, 1H), 8.53-8.52 (m, 1H), 8.34-8.29 (m, 2H), 8.14-8.09 (m, 1H), 7.89-7.81 (m, 2H), 7.42-7.38 (m, 1H). ESIMS found for C18H12FN5O m/z 334.0 (M+H).


Example 5

Preparation of N-(pyridin-3-yl)-5-(5-(trifluoromethyl)pyridin-yl)-1H-indazole-3-carboxamide (16) is depicted below in Scheme 31.




embedded image



Step 1


Preparation of intermediate 5-bromo-N-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXXXV) was performed following the procedure listed in Scheme 19, Step 4. Light yellow solid (5.5 g, 13.7 mmol, 88% yield). ESIMS found for C18H17BrN4O2 m/z 401.1 (M79Br+H) and 403.1 (M81Br+H).


Steps 2-3


Preparation of intermediate N-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-5-(5-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (CXXXVIII) was performed following the procedure listed in Scheme 26, Steps 1-2. Tan solid (295 mg, 0.63 mmol, 84% yield). ESIMS found for C24H20F3N5O2 m/z 468.1 (M+H).


Step 4


Preparation of N-(pyridin-3-yl)-5-(5-(trifluoromethyl)pyridin-yl)-1H-indazole-3-carboxamide (16) was performed following the procedure listed in Scheme 28, Step 4. White solid (95 mg, 0.25 mmol, 39.3% yield). 1H NMR (DMSO-d6) δ ppm 7.40 (dd, J=2.2 Hz, J=2 Hz, 1H), 7.84 (d, J=6.7 Hz, 1H), 7.93 (dd, J=1.5 Hz, J=7 Hz, 1H), 8.29-8.34 (m, 2H), 8.50 (s, 1H), 8.57 (s, 1H), 8.99 (s, 1H), 9.09 (d, J=2 Hz, 1H), 9.25 (d, J=1.6 Hz, 1H), 10.72 (brs, 1H); ESIMS found for C19H12F3N5O m/z 383.9 (M+H).


The following compounds were prepared in accordance with the procedure described in the above Example 5.




embedded image


5-(5-((Dimethylamino)methyl)pyridin-3-yl)-N-(6-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 26.


White solid (93 mg, 0.21 mmol, 78% yield). 1H NMR (DMSO-d6) δ ppm 2.24 (s, 6H), 3.57 (s, 2H), 7.86 (Abq, J=8 Hz, 2H), 7.93 (d, J=9 Hz, 1H), 8.04 (brs, 1H), 8.50 (d, J=7 Hz, 1H), 8.63 (dd, J=9 Hz, J=2 Hz, 1H), 8.85 (d, J=2 Hz, 1H), 9.27 (d, J=2 Hz, 1H), 11.11 (s, 1H), 14.11 (s, 1H); ESIMS found for C22H19F3N6O m/z 441.0 (M+H)




embedded image


N-(5-(3-(Pyridin-3-ylcarbamoyl)-1H-indazol-5-yl)pyridin-3-yl) morpholine-4-carboxamide 32.


White solid (132 mg, 0.30 mmol, 56% yield). 1H NMR (DMSO-d6) δ ppm 3.49 (t, J=5 Hz, 4H), 3.64 (t, J=5 Hz, 4H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.82 (d, J=1 Hz, 1H), 8.26 (t, J=2 Hz, 1H), 8.30-8.34 (m, 2H), 8.47 (s, 1H), 8.54 (d, J=2 Hz, 1H), 8.72 (d, J=2 Hz, 1H), 8.87 (s, 1H), 9.09 (d, 2 Hz, 1H), 10.71 (s, 1H), 14.01 (s, 1H); ESIMS found for C23H21N7O3 m/z 444.3 (M+H).




embedded image


5-(5-((Dimethylamino)methyl)pyridin-3-yl)-N-(6-(2-fluorophenoxy) pyridin-3-yl)-1H-indazole-3-carboxamide 36.


White solid (137 mg, 0.28 mmol, 53% yield). 1H NMR (DMSO-d6) δ ppm 2.20 (s, 6H), 3.53 (s, 2H), 7.16 (d, J=9 Hz, 1H), 7.22-7.40 (m, 4H), 7.82 (d/Abq, J=9 Hz, J=1 Hz, 2H), 8.00 (t, J=2 Hz, 1H), 8.38 (dd, J=9 Hz, J=3 Hz, 1H), 8.47 (s, 1H), 8.49 (d, J=2 Hz, 1H), 8.55 (d, J=3 Hz, 1H), 8.83 (d, J=2 Hz, 1H), 10.67 (s, 1H), 13.97 (brs, 1H); ESIMS found for C27H23FN6O2 m/z 383.1 (M+H).




embedded image


5-(5-(Cyclopropanecarboxamido)pyridin-3-yl)-N-(6-(4-methylpiperazin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 38.


White solid (39 mg, 0.08 mmol, 61% yield). 1H NMR (DMSO-d6) δ ppm 0.83-0.90 (m, 4H), 1.80-1.86 (m, 1H), 2.25 (brs, 3H), 2.45 (brs, 4H), 3.45 (brs, 4H), 6.86 (d, J=9 Hz, 1H), 7.79 (d, J=1 Hz, 1H), 8.04 (dd, J=9 Hz, J=3 Hz, 1H), 8.42 (t, J=2 Hz, 1H), 8.46 (s, 1H), 8.60 (dd, J=10 Hz, J=3 Hz, 2H), 8.76 (d, J=2 Hz, 1H), 10.34 (s, 1H), 10.56 (s, 1H), 13.90 (s, 1H); ESIMS found for C27H27N8O2 m/z 497.4 (M+H).




embedded image


5-(5-(Cyclopropanecarboxamido)pyridin-3-yl)-N-(6-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 39.


White solid (128 mg, 0.27 mmol, 45% yield). 1H NMR (DMSO-d6) δ ppm 0.82-0.90 (m, 4H), 1.80-1.86 (m, 1H), 7.84 (s, 2H0, 7.92 (d, J=9 Hz, 1H), 8.43 (d, J=2 Hz, 1H), 8.48 (s, 1H), 8.61-8.65 (m, 2H), 8.77 (d, J=2 Hz, 1H), 9.27 (d, J=2 Hz, 1H), 10.57 (s, 1H), 11.11 (s, 1H), 14.11 (s, 1H); ESIMS found for C23H17F3N6O2 m/z 467.1 (M+H).




embedded image


5-(5-((Dimethylamino)methyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 40.


White solid (312 mg, 0.84 mmol, 77% yield). 1H NMR (DMSO-d6) δ ppm 2.21 (s, 6H), 3.53 (s, 2H), 7.40 (dd, J=8 Hz, J=5 Hz. 1H), 7.83 (d/Abq, J=9 Hz., J=2 Hz. 2H), 8.01 (t, J=2 Hz, 1H), 8.29-8.34 (m, 2H), 8.48 (dd, J=4 Hz, J=1 Hz, 1H), 8.83 (d, J=2 Hz, 1H), 9.08 (d, J=3 Hz, 1H), 10.70 (s, 1H), 13.99 (brs, 1H); ESIMS found for C21H20N6O m/z 373.0 (M+H).




embedded image


5-(5-(Cyclopropanecarboxamido)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 41.


White solid (148 mg, 0.37 mmol, 71% yield). 1H NMR (DMSO-d6) δ ppm 0.83-0.90 (m, 4H), 1.80-1.87 (m, 1H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.82 (d, J=1 Hz, 1H), 8.29-8.34 (m, 2H), 8.43 (t, J=2 Hz, 1H), 8.47 (s, 1H), 8.62 (d, J=2 Hz, 1H), 8.76 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.57 (s, 1H), 10.70 (s, 1H), 14.01 (s, 1H); ESIMS found for C22H18N6O2 m/z 399.0 (M+H).




embedded image


N-(Pyridin-3-yl)-5-(5-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 43.


White solid (157 mg, 0.39 mmol, 76% yield). 1H NMR (DMSO-d6) δ ppm 1.70-1.74 (m, 4H), 2.46-2.52 (m, 4H), 3.71 (s, 2H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.83 (d/Abq, J=9 Hz, J=2 Hz, 2H), 8.02 (t, J=2 Hz, 1H), 8.29-8.34 (m, 2H), 8.48 (s, 1H), 8.51 (d, J=2 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.70 (s, 1H), 14.00 (s, 1H); ESIMS found for C23H22N6O m/z 399.0 (M+H).




embedded image


N-(6-Ethoxypyridin-3-yl)-5-(5-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 44.


White solid (62 mg, 0.14 mmol, 39% yield). 1H NMR (DMSO-d6) δ ppm 1.32 (t, J=7 Hz, 3H), 1.70-1.74 (m, 4H), 2.47-2.52 (m, 4H), 3.71 (s, 2H), 4.29 (q, J=7 Hz, 2H), 6.81 (d, J=9 Hz, 1H), 7.82 (d/Abq, J=9 Hz, J=2 Hz. 2H), 8.01 (t, J=2 Hz, 1H), 8.16 (dd, J=9 Hz, J=3 Hz, 1H), 8.46 (s, 1H), 8.51 (d, J=2 Hz, 1H), 8.63 (d, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.51 (s, 1H), 13.94 (brs, 1H); ESIMS found for C25H26N6O2 m/z 443.4 (M+H).




embedded image


N-(6-Ethoxypyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 45.


White solid (98 mg, 0.21 mmol, 44% yield). 1H NMR (DMSO-d6) δ ppm 1.32 (t, J=7 Hz, 3H), 1.34-1.42 (m, 2H), 1.47-1.53 (m, 4H), 2.38 (brs, 4H), 3.56 (s, 2H), 4.29 (q, J=7 Hz, 2H), 6.81 (d, J=9 Hz, 1H), 7.81 (d/Abq, J=9 Hz, J=2 Hz, 2H), 7.99 (t, J=2 Hz, 1H), 8.16 (dd, J=9 Hz, J=3 Hz, 1H), 8.46 (d, J=1 Hz, 1H), 8.49 (d, J=2 Hz, 1H), 8.63 (d, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.51 (s, 1H), 13.92 (brs, 1H); ESIMS found for C26H28N6O2 m/z 457.3 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 46.


White solid (126 mg, 0.31 mmol, 52% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.42 (m, 2H), 1.48-1.55 (m, 4H), 2.39 (brs, 4H), 3.57 (s, 2H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.83 (d/Abq, J=9 Hz, J=2 Hz, 2H), 7.99 (t, J=2 Hz, 1H), 8.30-8.34 (m, 2H), 8.48 (d, J=1 Hz, 1H), 8.49 (d, J=2 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.70 (s, 1H), 14.00 (brs, 1H); ESIMS found for C24H24N6O m/z 413.0 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(4-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 47.


White solid (150 mg, 0.31 mmol, 71% yield). 1H NMR (DMSO-d6) δ ppm 1.34-1.42 (m, 2H), 1.46-1.53 (m, 4H), 2.37 (brs, 4H), 3.55 (s, 2H), 7.81-7.87 (m, 3H), 7.98 (s, 1H), 8.41 (s, 1H), 8.48 (d, J=2 Hz, 1H), 8.75 (d, J=5 Hz, 1H), 8.80 (d, J=2 Hz, 1H), 9.07 (s, 1H), 10.22 (s, 1H), 14.06 (brs, 1H); ESIMS found for C25H23F3N6O m/z 481.0 (M+H).




embedded image


N-(Pyridin-3-yl)-5-(3-(pyrrolidin-1-ylmethyl)phenyl)-1H-indazole-3-carboxamide 49.


Tan amorphous solid (53.4 mg, 0.13 mmol, 72% yield). 1H NMR (DMSO-d6) δ ppm 1.70-1.71 (m, 4H), 2.47-2.49 (m, 4H), 3.67 (s, 2H), 7.31 (d, J=8 Hz, 1H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.44 (t, J=8 Hz, 1H), 7.58-7.60 (m, 1H), 7.63-7.64 (m, 1H), 7.76-7.78 (m, 2H), 8.30-8.34 (m, 2H), 8.44 (s, 1H), 9.08 (d, J=2 Hz, 1H), 10.68 (s, 1H), 13.93 (s, 1H); ESIMS found for C24H23N5O m/z 398 (M+H).




embedded image


N-(6-Phenylpyridin-3-yl)-5-(5-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 50.


Tan flaky solid (61.3 mg, 0.13 mmol, 74% yield). 1H NMR (DMSO-d6) δ ppm 1.71-1.72 (m, 4H), 3.72 (s, 2H), 7.39-7.42 (m, 1H), 7.47-7.50 (m, 2H), 7.81-7.86 (m, 2H), 8.00 (d, J=9 Hz, 1H), 8.02-8.03 (m, 1H), 8.08-8.10 (m, 2H), 8.45 (dd, J=9 Hz, J=3 Hz, 1H), 8.49-8.50 (m, 1H), 8.51 (d, J=2 Hz, 1H), 8.83 (d, J=2 Hz, 1H), 9.18 (d, J=3 Hz, 1H), 10.81 (s, 1H), 14.03 (s, 1H); ESIMS found for C29H26N6O m/z 475 (M+H).




embedded image


N-(Pyridin-3-yl)-5-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 51.


Yellow solid (32 mg, 0.08 mmol, 37.8% yield). 1H NMR (DMSO-d6) δ ppm 1.94-2.01 (m, 4H), 3.42-3.48 (m, 4H), 6.57 (d, J=9 Hz, 1H), 7.40 (dd, J=8 Hz, J=5 Hz, 1H), 7.72 (d, J=1 Hz, 2H), 7.85 (dd, J=9 Hz, J=3 Hz, 1H), 8.29-8.34 (m, 3H), 8.43 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.63 (s, 1H), 13.87 (s, 1H); ESIMS found for C22H20N6O m/z 385.0 (M+H).




embedded image


N-(6-Cyanopyridin-3-yl)-5-(5-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 52.


Beige solid (52 mg, 0.12 mmol, 49.1% yield). 1H NMR (DMSO-d6) δ ppm 1.70-1.75 (m, 4H), 3.31-3.36 (m, 4H), 7.85 (dq, J=9 Hz, J=2 Hz, 2H), 8.02 (s, 1H), 8.05 (d, J=9 Hz, 1H), 8.47 (s, 1H), 8.52 (d, J=2 Hz, 1H), 8.58 (dd, J=9 Hz, J=3 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.28 (d, J=2 Hz, 1H), 11.18 (s, 1H), 14.13 (brs, 1H); ESIMS found for C24H21N7O m/z 424.3 (M+H).




embedded image


5-(6-Methoxypyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 54.


White solid (79.7 mg, 0.23 mmol, 44.2% yield). 1H NMR (DMSO-d6) δ ppm 3.91 (s, 3H), 6.95 (d, J=9 Hz, 1H), 7.40 (dd, J=9 Hz, J=5 Hz, 1H), 7.78 (dd, J=11 Hz, J=2 Hz, 2H), 8.06 (dd, J=9 Hz, J=3 Hz, 1H), 8.29-8.34 (m, 2H), 8.39 (s, 1H), 8.51 (d, J=2 Hz, 1H), 9.08 (d, J=3 Hz, 1H), 10.67 (s, 1H), 13.91 (brs, 1H); ESIMS found for C19H15N5O2 m/z 346.0 (M+H).




embedded image


5-(5-Benzylpyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 55.


Yellow solid (101.9 mg, 0.25 mmol, 76% yield). 1H NMR (DMSO-d6) δ ppm 4.09 (s, 2H), 7.19-7.23 (m, 1H), 7.30-7.35 (m, 4H), 7.39-7.41 (m, 1H), 7.78-7.82 (m, 2H), 7.99 (t, J=2 Hz, 1H), 8.31-8.33 (m, 2H), 8.45 (s, 1H), 8.51 (d, J=2 Hz, 1H), 8.76 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.69 (s, 1H), 14.00 (s, 1H); ESIMS found for C25H19N5O m/z 406 (M+H).




embedded image


5-(5-Phenoxypyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 56.


White solid (73.6 mg, 0.18 mmol, 75% yield). 1H NMR (DMSO-d6) δ ppm 7.17-7.18 (m, 2H), 7.22-7.23 (m, 1H), 7.38-7.41 (m, 1H), 7.44-7.47 (m, 2H), 7.72-7.73 (m, 1H), 7.80-7.81 (m, 2H), 8.29-8.31 (m, 2H), 8.37-8.38 (m, 1H), 8.44-8.45 (m, 1H), 8.74 (d, J=2 Hz, 1H), 9.06 (d, J=2 Hz, 1H), 10.69 (s, 1H), 14.00 (s, 1H); ESIMS found for C24H17N5O2 m/z 408 (M+H).




embedded image


5-(5-(Pyrrolidin-1-ylmethyl)pyridin-3-yl)-N-(4-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 57.


White solid (64 mg, 0.14 mmol, 35.2% yield). 1H NMR (DMSO-d6) δ ppm 1.67-1.74 (m, 4H), 2.44-2.52 (m, 4H), 3.70 (s, 2H), 7.81-7.88 (m, 3H), 8.00 (d, J=2 Hz, 1H), 8.41 (s, 1H), 8.50 (d, J=2 Hz, 1H), 8.75 (d, J=5 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 9.07 (s, 1H), 10.22 (s, 1H), 14.01 (brs, 1H); ESIMS found for C24H21F3N6O m/z 467.3 (M+H).




embedded image


5-(5-(Cyclohexylcarbamoyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 59.


Light brown solid (117 mg, 0.27 mmol, 49.7% yield). 1H NMR (DMSO-d6) δ ppm 1.10-1.21 (m, 1H), 1.28-1.39 (m, 4H), 1.63 (d, J=12 Hz, 1H), 1.72-1.78 (m, 2H), 1.86-1.91 (m, 2H), 3.77-3.87 (m, 1H), 7.41 (dd, J=8 Hz, J=5 Hz, 1H), 7.84 (d, J=8 Hz, 1H), 7.91 (d, J=9 Hz, 1H), 8.30-8.36 (m, 2H), 8.48 (t, J=2 Hz, 1H), 8.55 (s, 1H), 8.59 (d, J=8 Hz, 1H), 8.99 (d, J=2 Hz, 1H), 9.04 (d, J=2 Hz, 1H), 9.09 (d, J=2 Hz, 1H), 10.72 (s, 1H), 14.04 (s, 1H); ESIMS found for C25H24N6O2 m/z 441.0 (M+H).




embedded image


5-(3-Fluoro-5-((4-methylpiperazin-1-yl)methyl)phenyl)-N-(4-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 60.


White solid (43 mg, 0.08 mmol, 76.3% yield). 1H NMR (DMSO-d6) δ ppm 1.23 (s, 3H), 2.22-2.50 (m, 8H), 3.56 (s, 2H), 7.12 (d, J=9 Hz, 1H), 7.42 (dd, J=8 Hz, J=2 Hz, 1H), 7.47 (s, 1H), 7.80 (d, J=1 Hz, 2H), 7.85 (d, J=5 Hz, 1H), 8.39 (s, 1H), 8.75 (d, J=5 Hz, 1H), 9.08 (s, 1H), 10.22 (s, 1H), 14.02 (brs, 1H); ESIMS found for C26H24F4N6O m/z 513.3 (M+H).




embedded image


5-(5-((4-Methylpiperazin-1-yl)methyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 61.


White solid (81.6 mg, 0.19 mmol, 55% yield). 1H NMR (DMSO-d6) δ ppm 2.14 (s, 3H), 2.33-2.42 (m, 8H), 3.60 (s, 2H), 7.39-7.41 (m, 1H), 7.81-7.85 (m, 2H), 8.00-8.01 (m, 1H), 8.31-8.33 (m, 2H), 8.47-8.48 (m, 1H), 8.49 (d, J=2 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.08 (d, J=3 Hz, 1H), 10.74 (s, 1H), 14.00 (s, 1H); ESIMS found for C24H25N7O m/z 427.8 (M+H).




embedded image


N-(6-Cyanopyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 62.


Off-white solid (42 mg, 0.10 mmol, 36.9% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.42 (m, 2H), 1.47-1.54 (m, 4H), 2.38 (brs, 4H), 3.57 (s, 2H), 7.85 (d, J=1 Hz, 2H), 8.00 (t, J=2 Hz, 1H), 8.05 (d, J=9 Hz, 1H), 8.47 (d, J=1 Hz, 1H), 8.50 (d, J=2 Hz, 1H), 8.58 (dd, J=9 Hz, J=3 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.28 (d, J=2 Hz, 1H), 11.18 (s, 1H), 14.12 (brs, 1H); ESIMS found for C25H23N7O m/z 438.1 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(6-(trifluoromethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 63.


White solid (78 mg, 0.16 mmol, 49% yield). 1H NMR (DMSO-d6) δ ppm 1.35-1.44 (m, 2H), 1.46-1.57 (m, 4H), 2.40 (brs, 4H), 3.59 (brs, 2H), 7.85 (s, 2H), 7.93 (d, J=9 Hz, 1H), 8.01 (s, 1H), 8.48 (s, 1H), 8.50 (s, 1H), 8.63 (d, J=8 Hz, 1H), 8.83 (s, 1H), 9.27 (s, 1H), 11.11 (s, 1H), 14.11 (brs, 1H); ESIMS found for C25H23F3N6O m/z 481.1 (M+H).




embedded image


5-(5-(Morpholinomethyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 64.


White solid (77 mg, 0.19 mmol, 66% yield). 1H NMR (DMSO-d6) δ ppm 2.41-2.43 (m, 4H), 3.58-3.60 (m, 4H), 3.61 (s, 2H), 7.39-7.41 (m, 1H), 7.81-7.85 (m, 2H), 8.02-8.03 (m, 1H), 8.31-8.33 (m, 2H), 8.47-8.48 (m, 1H), 8.51 (d, J=2 Hz, 1H), 8.83 (d, J=2 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.70 (s, 1H), 14.00 (s, 1H); ESIMS found for C23H22N6O2 m/z 415 (M+H).




embedded image


N-(6-Phenylpyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 65.


White solid (61.5 mg, 0.13 mmol, 68% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.53 (m, 4H), 2.38-2.39 (m, 4H), 3.57 (s, 2H), 7.39-7.43 (m, 1H), 7.47-7.50 (m, 2H), 7.82-7.86 (m, 2H), 7.99-8.01 (m, 2H), 8.08-8.10 (m, 2H), 8.44 (dd, J=9 Hz, J=3 Hz, 1H), 8.50-8.51 (m, 2H), 8.83 (d, J=2 Hz, 1H), 9.18 (d, J=3 Hz, 1H), 10.81 (s, 1H), 14.02 (s, 1H); ESIMS found for C30H28N6O m/z 489 (M+H).




embedded image


5-(5-Cyanopyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 66.


Beige solid (107 mg, 0.31 mmol, 66.7% yield). 1H NMR (DMSO-d6) δ ppm 7.40 (dd, J=8 Hz, J=4 Hz, 1H), 7.84 (d, J=8 Hz, 1H), 7.91 (dd, J=9 Hz, J=2 Hz, 1H), 8.30-8.34 (m, 2H), 8.57 (s, 1H), 8.72 (t, J=2 Hz, 1H), 9.03 (d, J=2 Hz, 1H), 9.09 (d, J=2 Hz, 1H), 9.23 (d, J=2 Hz, 1H), 10.72 (s, 1H), 14.06 (s, 1H); ESIMS found for C19H12N6O m/z 340.8 (M+H).




embedded image


5-(3-Fluoro-5-(piperidin-1-ylmethyl)phenyl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 67.


Yellow solid (84 mg, 0.20 mmol, 66% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.39 (m, 2H), 1.49-1.54 (m, 4H), 2.37-2.38 (m, 4H), 3.54 (s, 2H), 7.12-7.13 (m, 1H), 7.39-7.43 (m, 2H), 7.47-7.48 (m, 1H), 7.77-7.81 (m, 2H), 8.31-8.33 (m, 2H), 8.44-8.45 (m, 1H), 9.08 (d, J=2 Hz, 1H), 10.69 (s, 1H), 13.97 (s, 1H); ESIMS found for C25H24FN5O m/z 430 (M+H).




embedded image


5-(5-(Morpholinomethyl)pyridin-3-yl)-N-(6-(trifluoromethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 68.


White solid (72 mg, 0.15 mmol, 30.5% yield). 1H NMR (DMSO-d6) δ ppm 2.43 (brs, 4H), 3.56-3.63 (m, 4H), 3.62 (s, 2H), 7.85 (Abq, J=9 Hz, 2H), 7.93 (d, J=9 Hz, 1H), 8.04 (s, 1H), 8.49 (s, 1H), 8.52 (d, J=1 Hz, 1H), 8.63 (dd, J=9 Hz, J=3 Hz, 1H), 8.84 (d, J=2 Hz, 1H), 9.27 (d, J=2 Hz, 1H), 11.11 (s, 1H), 14.11 (brs, 1H); ESIMS found for C24H21F3N6O2 m/z 483.3 (M+H).




embedded image


N-(6-Morpholinopyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 69.


Light yellow solid (58 mg, 0.12 mmol, 36.4% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.44 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.33-2.40 (m, 4H), 3.40 (t, J=5 Hz, 4H), 3.56 (s, 2H), 3.71 (t, 5 Hz, 4H), 6.89 (d, J=9 Hz, 1H), 7.78 (d, J=8 Hz, 1H), 7.81 (d, J=9 Hz, 1H), 7.97 (t, J=2 Hz, 1H), 8.06 (dd, J=9 Hz, J=2 Hz, 1H), 8.46 (d, J=10 Hz, 1H), 8.60 (d, J=2 Hz, 1H), 8.79 (d, J=2 Hz, 1H), 10.35 (s, 1H), 13.90 (brs, 1H); ESIMS found for C28H31N7O2 m/z 498.0 (M+H).




embedded image


N-(6-(4-Methylpiperazin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 70.


Light yellow solid (37 mg, 0.07 mmol, 39.2% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.44 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.22 (s, 3H), 2.35-2.42 (m, 8H), 3.44 (t, J=5 Hz, 4H), 3.56 (s, 2H), 6.86 (d, J=9 Hz, 1H), 7.79 (d, J=9 Hz, 1H), 7.82 (d, J=10 Hz, 1H), 7.98 (d, J=2 Hz, 1H), 8.03 (dd, J=9 Hz, J=3 Hz, 1H), 8.48 (d, J=11 Hz, 1H), 8.58 (d, J=3 Hz, 1H), 8.81 (d, J=3 Hz, 1H), 10.34 (s, 1H), 13.89 (brs, 1H); ESIMS found for C29H34N8O m/z 511.5 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(6-(pyrrolidin-1-yl) pyridin-3-yl)-1H-indazole-3-carboxamide 71.


Tan solid (53.9 mg, 0.11 mmol, 53% yield). 1H NMR (DMSO-d6) δ ppm 1.38-1.39 (m, 2H), 1.51-1.52 (m, 4H), 1.93-1.96 (m, 4H), 2.36-2.38 (m, 4H), 3.36-3.39 (m, 4H), 3.56 (s, 2H), 6.46 (d, J=9 Hz, 1H), 7.78-7.83 (m, 2H), 7.96 (dd, J=9 Hz, J=3 Hz, 1H), 7.98-7.99 (m, 1H), 8.46-8.47 (m, 2H), 8.49 (d, J=3 Hz, 1H), 8.80-8.81 (m, 1H), 10.23 (s, 1H), 13.87 (s, 1H); ESIMS found for C28H31N7O m/z 482 (M+H).




embedded image


N-(6-(3-Fluorophenyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 72.


White solid (54.8 mg, 0.11 mmol, 64% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.50-1.54 (m, 4H), 2.38-2.39 (m, 4H), 3.57 (s, 2H), 7.22-7.26 (m, 1H), 7.51-7.55 (m, 1H), 7.82-7.86 (m, 2H), 7.88-7.91 (m, 1H), 7.94-7.96 (m, 1H), 8.00-8.01 (m, 1H), 8.06 (d, J=9 Hz, 1H), 8.46 (dd, J=9 Hz, J=3 Hz, 1H), 8.50 (s, 2H), 8.82 (d, J=2 Hz, 1H), 9.20 (d, J=2 Hz, 1H), 10.86 (s, 1H), 14.03 (s, 1H); ESIMS found for C30H27FN6O m/z 507 (M+H).




embedded image


N-(6-(4-Fluorophenyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 73.


White solid (50.8 mg, 0.10 mmol, 55% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.53 (m, 4H), 2.36-2.39 (m, 4H), 3.57 (s, 2H), 7.29-7.32 (m, 2H), 7.82-7.86 (m, 2H), 7.98-8.01 (m, 2H), 8.12-8.15 (m, 2H), 8.43 (dd, J=9 Hz, J=3 Hz, 1H), 8.49 (s, 2H), 8.82 (d, J=2 Hz, 1H), 9.17 (d, J=3 Hz, 1H), 10.81 (s, 1H), 14.02 (s, 1H); ESIMS found for C30H27FN6O m/z 507 (M+H).




embedded image


N-(6-((2-(Dimethylamino)ethyl)(methyl)amino)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 74.


Light yellow solid (88.5 mg, 0.17 mmol, 61.7% yield). 1H NMR (DMSO-d6) δ ppm 1.38-1.42 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.18 (s, 6H), 2.34-2.40 (m, 6H), 2.99 (s, 3H), 3.56 (s, 2H), 3.61 (t, J=7 Hz, 2H), 6.61 (d, J=9 Hz, 1H), 7.79 (d, J=9 Hz, 1H), 7.81 (d, J=9 Hz, 1H), 7.95 (dd, J=9 Hz, J=3 Hz, 1H), 7.98 (t, J=2 Hz, 1H), 8.46 (s, 1H), 8.48 (d, J=2 Hz, 2H), 8.81 (d, J=2 Hz, 1H), 10.24 (s, 1H), 13.84 (brs, 1H); ESIMS found for C29H36N8O m/z 513.5 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(pyridazin-4-yl)-1H-indazole-3-carboxamide 75.


White solid (53 mg, 0.13 mmol, 33.7% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.47-1.54 (m, 4H), 2.33-2.42 (m, 4H), 3.57 (s, 2H), 7.85 (s, 2H), 8.00 (t, J=2 Hz, 1H), 8.25 (dd, J=6 Hz, J=3 Hz, 1H), 8.47 (t, J=1 Hz, 1H), 8.50 (d, J=2 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 9.09 (d, J=6 Hz, 1H), 9.71 (dd, J=3 Hz, J=1 Hz, 1H), 11.16 (s, 1H), 14.16 (brs, 1H); ESIMS found for C23H23N7O m/z 414.1 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(pyridin-3-ylmethyl)-1H-indazole-3-carboxamide 76.


White solid (26.8 mg, 0.06 mmol, 27% yield). 1H NMR (DMSO-d6) δ ppm 1.38-1.39 (m, 2H), 1.49-1.51 (m, 4H), 2.36-2.37 (m, 4H), 3.55 (s, 2H), 4.53 (d, J=6 Hz, 2H), 7.35 (dd, J=8 Hz, J=5 Hz, 1H), 7.74-7.80 (m, 3H), 7.95-7.96 (m, 1H), 8.41-8.42 (m, 1H), 8.45-8.46 (m, 1H), 8.48-8.49 (m, 1H), 8.58-8.59 (m, 1H), 8.78 (d, J=2 Hz, 1H), 9.17 (t, J=6 Hz, 1H), 13.77 (s, 1H); ESIMS found for C25H26N6O m/z 427 (M+H).




embedded image


N-Cyclohexyl-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 77.


White solid (50.4 mg, 0.12 mmol, 72.5% yield). 1H NMR (DMSO-d6) δ ppm 1.12-1.47 (m, 7H), 1.50-1.53 (m, 4H), 1.60-1.63 (m, 1H), 1.73-1.75 (m, 2H), 1.83-1.84 (m, 2H), 2.37-2.38 (m, 4H), 3.55 (s, 2H), 3.81-3.87 (m, 1H), 7.73-7.78 (m, 2H), 7.95-7.96 (m, 1H), 8.14 (d, J=8 Hz, 1H), 8.41-8.42 (m, 1H), 8.47 (d, J=2 Hz, 1H), 8.78 (d, J=2 Hz, 1H), 13.67 (s, 1H); ESIMS found for C25H31N5O m/z 418 (M+H).




embedded image


N-(Benzo[d][1,3]dioxol-5-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 78.


White solid (48.6 mg, 0.11 mmol, 22.1% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.43 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.36-2.42 (m, 4H), 3.56 (s, 2H), 6.01 (s, 2H), 6.90 (d, J=9 Hz, 1H), 7.37 (dd, J=9 Hz, J=2 Hz, 1H), 7.57 (d, J=2 Hz, 1H), 7.91 (dd, J=9 Hz, J=Hz, 1H), 7.82 (dd, J=9 Hz, J=1 Hz, 1H), 7.99 (t, J=2 Hz, 1H), 8.47 (dd, J=12 Hz, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.34 (s, 1H), 13.89 (s, 1H); ESIMS found for C26H25N5O3 m/z 456.0 (M+H).




embedded image


N-(2,3-Dihydrobenzo[b][1,4]dioxin-6-yl)-5-(5-(piperidin-1-ylmethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 79.


White solid (98.4 mg, 0.21 mmol, 38.7% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.42 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.34-2.41 (m, 4H), 3.56 (s, 2H), 4.20-4.27 (m, 4H), 6.82 (d, J=9 Hz, 1H), 7.35 (dd, J=9 Hz, J=3 Hz, 1H), 7.51 (d, J=3 Hz, 1H), 7.78 (dd, J=9 Hz, J=1 Hz, 1H), 7.81 (dd, J=9 Hz, J=1 Hz, 1H), 7.98 (t, J=2 Hz, 1H), 8.47 (dd, J=12 Hz, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.26 (s, 1H), 13.87 (brs, 1H); ESIMS found for C27H27N5O3 m/z 470.4 (M+H).




embedded image


N-(5-Benzylpyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 80.


White solid (81.9 mg, 0.16 mmol, 59% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.41 (m, 2H), 1.49-1.53 (m, 4H), 2.37-2.39 (m, 4H), 3.56 (s, 2H), 4.00 (s, 2H), 7.20-7.23 (m, 1H), 7.28-7.34 (m, 4H), 7.79-7.84 (m, 2H), 7.98-7.99 (m, 1H), 8.23-8.24 (m, 1H), 8.25 (d, J=2 Hz, 1H), 8.45-8.46 (m, 1H), 8.49 (d, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 8.89 (d, J=2 Hz, 1H), 10.65 (s, 1H), 13.97 (s, 1H); ESIMS found for C31H30N6O m/z 503 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(pyrazin-2-yl)-1H-indazole-3-carboxamide 81.


White solid (104 mg, 0.25 mmol, 41.7% yield). 1H NMR (DMSO-d6) δ ppm 1.35-1.42 (m, 2H), 1.51 (quin, J=5 Hz, 4H), 2.33-2.42 (m, 4H), 3.57 (s, 2H), 7.83 (d, J=9 Hz, 1H), 7.85 (d, J=9 Hz, 1H), 8.00 (s, 1H), 8.45 (d, J=2 Hz, 1H), 8.46 (s, 1H), 8.50 (s, 1H), 8.83 (d, J=2 Hz, 1H), 9.50 (s, 1H), 10.36 (s, 1H), 14.11 (brs, 1H); ESIMS found for C23H23N7O m/z 413.9 (M+H).




embedded image


N-Phenyl-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 82.


White solid (97.8 mg, 0.24 mmol, 81% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.53 (m, 4H), 2.37-2.39 (m, 4H), 3.57 (s, 2H), 7.09-7.12 (m, 1H), 7.34-7.37 (m, 2H), 7.80 (d, J=9 Hz, 1H), 7.83 (dd, J=9 Hz, 2 Hz, 1H), 7.907.92 (m, 2H), 7.99-8.00 (m, 1H), 8.47-8.48 (m, 1H), 8.49 (d, J=2 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.40 (s, 1H), 13.92 (s, 1H); ESIMS found for C25H25N5O m/z 412 (M+H).




embedded image


(4-Methylpiperazin-1-yl)(5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazol-3-yl)methanone 83.


Light yellow amorphous solid (74.6 mg, 0.18 mmol, 93% yield). 1H NMR (DMSO-d6) δ ppm 1.38-1.39 (m, 2H), 1.48-1.53 (m, 4H), 2.22 (s, 3H), 2.36-2.41 (m, 8H), 3.55 (s, 2H), 3.72-3.73 (m, 2H), 4.01-4.02 (m, 2H), 7.73 (d, J=9 Hz, 1H), 7.79 (dd, J=9 Hz, J=2 Hz, 1H), 7.95-7.96 (m, 1H), 8.22 (d, J=1 Hz, 1H), 8.46 (d, J=2 Hz, 1H), 8.78 (d, J=2 Hz, 1H), 13.64 (s, 1H); ESIMS found for C24H30N6O m/z 419 (M+H).




embedded image


5-(5-(((2R,6 S)-2,6-Dimethylpiperidin-1-yl)methyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 84.


Beige solid (76.5 mg, 0.17 mmol, 75.5% yield). 1H NMR (DMSO-d6) δ ppm 1.00 (d, J=6 Hz, 6H), 1.21-1.35 (m, 3H), 1.55 (d, J=11 Hz, 2H), 1.60-1.65 (m, 1H), 2.45-2.53 (m, 2H), 3.84 (s, 1H), 7.40 (dd, J=7 Hz, 3 Hz, 1H), 7.79 (dd, J=9 Hz, J=2 Hz, 1H), 7.83 (dd, J=9 Hz, J=1 Hz, 1H), 8.04 (s, 1H), 8.29-8.35 (m, 2H), 8.46 (s, 1H), 8.60 (d, J=2 Hz, 1H), 8.73 (d, J=2 Hz, 1H), 9.08 (d, J=3 Hz, 1H), 10.70 (s, 1H), 14.00 (brs, 1H); ESIMS found for C26H28N6O m/z 441.3 (M+H).




embedded image


N-(5-((Dimethylamino)methyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 86.


White solid (41.5 mg, 0.09 mmol, 72% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.54 (m, 4H), 2.19 (s, 6H), 2.36-2.39 (m, 4H), 3.44 (s, 2H), 3.58 (s, 2H), 7.81 (d, J=9 Hz, 1H), 7.85 (dd, J=9 Hz, J=2 Hz, 1H), 8.00-8.01 (m, 1H), 8.21 (d, J=2 Hz, 1H), 8.37-8.38 (m, 1H), 8.49-8.50 (m, 2H), 8.83 (d, J=2 Hz, 1H), 8.91 (d, J=2 Hz, 1H), 10.69 (s, 1H), 14.01 (brs, 1H); ESIMS found for C27H31N7O m/z 470 (M+H).




embedded image


N-(1-Methylpiperidin-4-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 87.


White amorphous solid (18.2 mg, 0.04 mmol, 59.8% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.53 (m, 4H), 1.66-1.75 (m, 4H), 1.95-2.00 (m, 2H), 2.18 (s, 3H), 2.37-2.38 (m, 4H), 2.77 (d, J=11 Hz, 2H), 3.55 (s, 2H), 3.81-3.83 (m, 1H), 7.73-7.75 (m, 1H), 7.77-7.79 (m, 1H), 7.95-7.96 (m, 1H), 8.25 (d, J=8 Hz, 1H), 8.41-8.42 (m, 1H), 8.47 (d, J=2 Hz, 1H), 8.78 (d, J=2 Hz, 1H), 13.70 (s, 1H); ESIMS found for C25H32N6O m/z 433 (M+H).




embedded image


N-(5-((Dimethylamino)methyl)pyridin-3-yl)-5-(5-(pyrrolidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 106.


White solid (39.4 mg, 0.09 mmol, 74% yield). 1H NMR (DMSO-d6) δ ppm 1.71-1.73 (m, 4H), 2.49-2.50 (m, 4H), 2.18 (s, 6H), 3.43 (s, 2H), 3.71 (s, 2H), 7.81 (d, J=9 Hz, 1H), 7.84 (ABq, J=9 Hz, 1H), 8.02-8.03 (m, 1H), 8.21 (d, J=2 Hz, 1H), 8.37-8.38 (m, 1H), 8.48-8.49 (m, 1H), 8.51 (d, J=2 Hz, 1H), 8.83 (d, J=2 Hz, 1H), 8.91 (d, J=2 Hz, 1H), 10.68 (s, 1H), 13.98 (s, 1H); ESIMS found for C26H29N7O m/z 456 (M+H).




embedded image


N-(5-((4-Methylpiperazin-1-yl)methyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 124.




embedded image


N-(6-(Piperidin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 126.


Grey solid (92.7 mg, 0.19 mmol, 29.0% yield). 1H NMR (DMSO-d6) δ ppm 1.48-1.64 (m, 12H), 2.32-2.43 (m, 4H), 3.48 (t, J=4.5 Hz, 4H), 3.56 (s, 2H), 6.83 (d, J=9 Hz, 1H), 7.80 (ABq, J=10 Hz, 2H), 7.98 (s, 1H), 8.00 (d, J=2.4 Hz, 1H), 8.47 (d, J=10 Hz, 2H), 8.55 (d, J=2.5 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.27 (s, 1H), 13.86 (s, 1H); ESIMS found for C29H33N7O m/z 496.5 (M+H).




embedded image


N-(3-Fluorophenyl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 162.


White solid (176 mg, 0.41 mmol, 56.8% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.47-1,55 (m, 4H), 2.38 (brs, 4H), 3.56 (s, 2H), 6.93 (dt, J=9 Hz, J=3 Hz, 1H), 7.39 (q, J=8 Hz, 1H), 7.75 (dd, J=8 Hz, J=1 Hz, 1H), 7.82 (d/Abq, J=9 Hz, J=1 Hz, 2H), 7.89 (td, J=12 Hz, J=2 Hz, 1H), 7.99 (t, J=2 Hz, 1H), 8.47 (s, 1H), 8.49 (d, J=2 Hz, 1H), 8.82 (d, J=2 Hz, 1H), 10.66 (s, 1H), 13.97 (brs, 1H); ESIMS found for C25H24FN5O m/z 430.0 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(tetrahydro-2H-pyran-4-yl)-1H-indazole-3-carboxamide 163.


Tan amorphous solid (88 mg, 0.21 mmol, 88% yield). 1H NMR (DMSO-d6) δ ppm 1.39-1.40 (m, 2H), 1.49-1.53 (m, 4H), 1.69-1.76 (m, 4H), 2.37-2.38 (m, 4H), 3.39-3.42 (m, 2H), 3.56 (s, 2H), 3.88-3.90 (m, 2H), 4.05-4.10 (m, 1H), 7.74 (d, J=9 Hz, 1H), 7.77-7.79 (m, 1H), 7.95-7.96 (m, 1H), 8.37 (d, J=8 Hz, 1H), 8.41-8.42 (m, 1H), 8.47 (d, J=2 Hz, 1H), 8.79 (d, J=2 Hz, 1H), 13.72 (s, 1H); ESIMS found for C24H29N5O2 m/z 420 (M+H).




embedded image


N-(5-Fluoropyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 168.


White solid (286 mg, 0.66 mmol, 56% yield). 1H NMR (DMSO-d6) δ ppm 1.39 (m, 2H), 1.49-1.53 (m, 4H), 2.38 (brs, 4H), 3.56 (s, 2H), 7.81-7.86 (m, 2H), 7.99 (s, 1H), 8.31-8.34 (m, 2H), 8.47 (s, 1H), 8.49 (d, J=1.3 Hz, 1H), 8.82 (d, J=1.7 Hz, 1H), 8.99 (s, 1H), 10.97 (s, 1H), 14.07 (brs, 1H); ESIMS found for C24H23FN6O m/z 431.4 (M+H).




embedded image


N-(6-(4-Hydroxypiperidin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 169.


Off-white solid (33 mg, 0.06 mmol, 53.8% yield). 1H NMR (DMSO-d6) δ ppm 1.32-1.43 (m, 4H), 1.45-1.57 (m, 4H), 1.74-1.83 (m, 2H), 2.33-2.44 (m, 4H), 3.04 (t, J=10 Hz, 2H), 3.56 (s, 2H), 3.63-3.73 (m, 1H), 3.93-4.02 (m, 2H), 4.72 (s, 1H), 6.85 (d, J=9 Hz, 1H), 7.80 (ABq, J=10 Hz, 2H), 7.99 (d, J=7 Hz, 2H), 8.47 (d, J=10 Hz, 2H), 8.54 (s, 1H), 8.81 (s, 1H), 10.28 (s, 1H), 13.87 (s, 1H); ESIMS found for C29H33N7O2 m/z 512.3 (M+H).




embedded image


5-(5-((4-Hydroxypiperidin-1-yl)methyl)pyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 170.


Off-white solid (125.4 mg, 0.25 mmol, 73.2% yield). 1H NMR (DMSO-d6) δ ppm 1.93-1.96 (m, 4H), 2.09-2.12 (m, 2H), 2.70-2.72 (m, 2H), 3.37-3.39 (m, 4H), 3.46-3.47 (m, 1H), 3.58 (s, 1H), 4.52 (d, J=4 Hz, 1H), 6.46 d, J=9 Hz, 1H), 7.77-7.82 (m, 2H), 7.95-7.98 (m, 2H), 8.44-8.48 (m, 2H), 8.49 (d, J=2.5 Hz, 1H), 8.80 (d, J=2.1 Hz, 1H), 10.20 (s, 1H), 13.85 (s, 1H); ESIMS found for C28H31N7O2 m/z 498 (M+H).




embedded image


N-(5-Methyl-6-(pyrrolidin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 172.


Off-white solid (186 mg, 0.38 mmol, 72.2% yield). 1H NMR (DMSO-d6) δ ppm 1.34-1.43 (m, 2H), 1.47-1.55 (m, 4H), 1.82-1.89 (m, 4H), 2.30 s, 3H), 2.33-2.42 (m, 4H), 3.43 (t, J=6.6 Hz, 4H), 3.56 (s, 2H), 7.81 (ABq, J=10 Hz, 2H), 7.89 (d, J=2 Hz, 1H), 7.98 (s, 1H), 8.38 (d, J=2 Hz, 1H), 8.47 (d, J=8 Hz, 2H), 8.81 (d, J=2 Hz, 1H), 10.24 (s, 1H), 13.86 (s, 1H); ESIMS found for C29H33N7O m/z 496.4 (M+H).




embedded image


N-(6-(Azetidin-1-yl)-5-methylpyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 173.


Off-white solid (184 mg, 0.38 mmol, 62.6% yield). 1H NMR (DMSO-d6) δ ppm 1.35-1.43 (m, 2H), 1.47-1.54 (m, 4H), 2.16 (s, 3H), 2.22 (quin, J=7 Hz, 2H), 2.34-2.42 (m, 4H), 3.56 (s, 2H), 4.00 (t, J=7 Hz, 4H), 7.81 (ABq, J=10 Hz, 2H), 7.85 (d, J=2 Hz, 1H), 8.39 (d, J=2 Hz, 1H), 8.47 (d, J=10 Hz, 2H), 8.81 (d, J=2 Hz, 1H), 10.24 (s, 1H), 13.87 (s, 1H); ESIMS found for C28H31N7O m/z 482.0 (M+H).




embedded image


N-(6-(Azetidin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 174.


White solid (14.9 mg, 0.03 mmol, 11.0% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.47-1.54 (m, 4H), 2.32 (quin, J=7 Hz, 2H), 2.35-2.42 (m, 4H), 3.56 (s, 2H), 3.92 (t, J=7 Hz, 4H), 6.39 (d, J=9 Hz, 1H), 7.77-7.83 (m, 2H), 7.98 (dd, J=9 Hz, J=2 Hz, 2H), 8.42-8.53 (m, 3H), 8.78-8.84 (m, 1H), 10.27 (s, 1H), 13.87 (s, 1H); ESIMS found for C27H29N7O m/z 468.0 (M+H).




embedded image


N-(6-Methoxypyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 175.


White solid (31.2 mg, 0.07 mmol, 25.8% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.47-1.55 (m, 4H), 2.33-2.42 (m, 4H), 3.56 (s, 2H), 3.85 (s, 3H), 6.84 (d, J=9 Hz, 1H), 7.81 (ABq, J=12 Hz, 2H), 7.98 (s, 1H), 8.18 (dd, J=9 Hz, J=2.7 Hz, 1H), 8.47 (dd, J=10 Hz, J=1 Hz, 2H), 8.65 (d, J=2.6 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.50 (s, 1H), 13.91 (brs, 1H); ESIMS found for C25H26N6O2 m/z 443.4 (M+H).




embedded image


N-(2-Aminopyrimidin-5-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 176.


Yellow solid (412 mg, 0.96 mmol, 52.5% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.43 (m, 2H), 1.47-1.54 (m, 4H), 2.35-2.41 (m, 4H), 3.56 (s, 2H), 6.49 (s, 2H), 7.81 (ABq, J=10 Hz, 2H), 7.98 (s, 1H), 8.47 (dd, J=12 Hz, J=2 Hz, 2H), 8.63 (s, 1H), 8.81 (d, J=2 Hz, 1H), 10.32 (s, 1H), 13.91 (s, 1H); ESIMS found for C23H24N8O m/z 429.3 (M+H).




embedded image


N-(6-(Piperazin-1-yl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 177.


Tan solid (160 mg, 0.32 mmol, 28.5% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.43 (m, 2H), 1.48-1.54 (m, 4H), 2.34-2.41 (m, 4H), 2.79 (t, J=5 Hz, 4H), 3.36 (t, J=5 Hz, 4H), 3.56 (s, 2H), 6.82 (d, J=9 Hz, 1H), 7.81 (ABq, J=10 Hz, 2H), 7.98 (s, 1H), 8.02 (dd, J=9 Hz, J=2.7 Hz, 1H), 8.47 (dd, J=9 Hz, J=2 Hz, 2H), 8.57 (d, J=2.5 Hz, 1H), 8.81 (d, J=2 Hz, 1H), 10.29 (s, 1H); ESIMS found for C28H32N8O m/z 497.1 (M+H).




embedded image


N-(6-Hydroxypyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 178.


Off-white solid (78.3 mg, 0.18 mmol, 52.4% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.48-1.54 (m, 4H), 2.35-2.42 (m, 4H), 3.56 (s, 2H), 6.38 (d, J=10 Hz, 1H), 7.80 (ABq, J=11 Hz, 2H), 7.83 (dd, J=10 Hz, J=3 Hz, 1H), 7.97 (s, 1H), 8.04 (d, J=2.5 Hz, 1H), 8.44 (s, 1H), 8.48 (d, J=2 Hz, 1H), 8.80 (d, J=2 Hz, 1H), 10.27 (s, 1H), 11.42 (brs, 1H), 13.87 (brs, 1H); ESIMS found for C24H24N6O2 m/z 429.1 (M+H).




embedded image


5-(5-(Piperidin-1-ylmethyl)pyridin-3-yl)-N-(6-(pyrrolidine-1-carbonyl)pyridin-3-yl)-1H-indazole-3-carboxamide 179.


Light yellow solid (61 mg, 0.12 mmol, 37.8% yield). 1H NMR (DMSO-d6) δ ppm 1.37-1.43 (m, 2H), 1.48-1.55 (m, 4H), 1.82-1.90 (m, 4H), 2.38 (brs, 4H), 3.17 (d, J=5 Hz, 2H), 3.51 (t, J=7 Hz, 2H), 3.57 (s, 2H), 3.70 (t, J=7 Hz, 2H), 7.79 (d, J=9 Hz, 1H), 7.84 (Abq, J=11 Hz, 2H), 8.00 (s, 1H), 8.46 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.48 (dd, J=9 Hz, J=2 Hz, 2H), 8.82 (d, J=2 Hz, 1H), 9.10 (d, J=2 Hz, 1H), 10.91 (s, 1H), 14.05 (brs, 1H); ESIMS found for C29H31N7O2 m/z 510.6 (M+H).




embedded image


N-(6-(Cyclopentylcarbamoyl)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 181.


Light yellow solid (18 mg, 0.04 mmol, 16.6% yield). 1H NMR (DMSO-d6) δ ppm 1.50-1.64 (m, 4H), 1.67-1.76 (m, 2H), 1.85-1.94 (m, 4H), 4.24 (quin, J=8 Hz, 1H), 7.53 (dd, J=8 Hz, J=5 Hz, 1H), 7.84 (ABq, 2H), 8.03 (d, J=9 Hz, 1H), 8.14 (d, J=8 Hz, 1H), 8.45 (d, J=8 Hz, 1H), 8.48 (s, 1H), 8.54 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.60 (d, J=4 Hz, 1H), 8.94 (d, J=2 Hz, 1H), 9.16 (d, J=2 Hz, 1H), 10.97 (s, 1H), 14.08 (brs, 1H); ESIMS found for C24H22N6O2 m/z 427.1 (M+H).




embedded image


5-(5-Aminopyridin-3-yl)-N-(6-(piperidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 182.


Off-white solid (23.4 mg, 0.06 mmol, 19.4% yield). 1H NMR (DMSO-d6) δ ppm 1.51-1.63 (m, 6H), 3.47 (t, J=5 Hz, 4H), 5.45 (s, 2H), 6.83 (d, J=10 Hz, 1H), 7.24 (t, J=2 Hz, 1H), 7.73 (dq, J=9 Hz, J=2 Hz, 2H), 7.94 (d, J=2.5 Hz, 1H), 8.00 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.08 (d, J=2 Hz, 1H), 8.40 (s, 1H), 8.56 (d, J=2.5 Hz, 1H), 10.27 (s, 1H), 13.84 (s, 1H); ESIMS found for C23H23N7O m/z 414.3 (M+H).




embedded image


5-(5-((3,3-Difluoropyrrolidin-1-yl)methyl)pyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 183.


Off-white solid (307 mg, 0.61 mmol, 39.6% yield). 1H NMR (DMSO-d6) δ ppm 1.95 (t, J=6.5 Hz, 4H), 2.28 (tt, J=13.5 Hz, J=7 Hz, 2H), 2.76 (t, J=7 Hz, 2H), 2.94 (t, J=13.5 Hz, 2H), 3.38 (t, J=6.5 Hz, 4H), 3.77 (s, 2H), 6.46 (d, J=9 Hz, 1H), 7.81 (dq, J=8.5 Hz, J=1.5 Hz, 2H), 7.97 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.03 (s, 1H), 8.48 (s, 1H), 8.49 (d, J=2.5 Hz, 1H), 8.52 (s, 1H), 8.84 (d, J=2 Hz, 1H), 10.23 (s, 1H), 13.87 (s, 1H); ESIMS found for C27H27F2N7O m/z 504.0 (M+H).




embedded image


N-(6-(Cyclopentylcarbamoyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide 184.


White solid (3.2 mg, 0.01 mmol, 18.5% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.43-1.64 (m, 8H), 1.64-1.76 (m, 2H), 1.82-1.93 (m, 2H), 2.38 (brs, 4H), 3.57 (s, 2H), 4.24 (quin, J=7 Hz, 1H), 7.84 (ABq, J=10 Hz, 2H), 8.00 (s, 1H), 8.03 (d, J=9 Hz, 1H), 8.44 (d, J=8 Hz, 1H), 8.48 (dd, J=8 Hz, J=2 Hz, 2H), 8.55 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.82 (d, J=2.5 Hz, 1H), 9.16 (d, J=2.5 Hz, 1H), 10.98 (s, 1H), 14.06 (brs, 1H); ESIMS found for C30H33N7O2 m/z 524.5 (M+H).




embedded image


N-(6-(Methylsulfonyl)pyridin-3-yl)-5-(5-(piperidin-1-ylmethyl) pyridin-3-yl)-1H-indazole-3-carboxamide 185.


White solid (72 mg, 0.15 mmol, 56.4% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.43 (m, 2H), 1.48-1.55 (m, 4H), 2.39 (brs, 4H), 3.27 (s, 3H), 3.57 (s, 2H), 7.85 (s, 2H), 8.00 (s, 1H), 8.08 (d, J=8.5 Hz, 1H), 8.49 (dd, J=10 Hz, J=1.5 Hz, 2H), 8.83 (d, J=2.5 Hz, 1H), 9.26 (d, J=2.5 Hz, 1H), 11.19 (s, 1H), 14.13 (brs, 1H); ESIMS found for C25H26N6O3S m/z 491.1 (M+H).




embedded image


5-(5-(4-Methylpiperazin-1-yl)pyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 186.


Off-white solid (196 mg, 0.41 mmol, 47.8% yield). 1H NMR (DMSO-d6) δ ppm 1.89-1.98 (m, 4H), 2.27 (brs, 3H), 3.25-3.42 (m, 12H), 6.45 (d, J=9 Hz, 1H), 7.53 (s, 1H), 7.77 (q, J=8.5 Hz, 2H), 7.96 (d, J=6.5 Hz, 1H), 8.31 (d, J=5.5 Hz, 2H), 8.43 (s, 1H), 8.48 (s, 1H), 10.21 (s, 1H), 13.83 (s, 1H); ESIMS found for C27H30N8O m/z 483.4 (M+H).




embedded image


5-(5-Morpholinopyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 187.


White solid (92 mg, 0.20 mmol, 43.5% yield). 1H NMR (DMSO-d6) δ ppm 1.94 (t, J=6.5 Hz, 4H), 3.28 (t, J=4.5 Hz, 4H), 3.38 (t, J=6.5 Hz, 4H), 3.78 (t, J=4.5 Hz, 4H), 6.45 (d, J=9 Hz, 1H), 7.55 (s, 1H), 7.77 (dq, J=8.5 Hz, J=1.5 Hz, 2H), 7.96 (dd, J=9 Hz, J=2.5 Hz 1H), 8.33 (dd, J=6.5 Hz, J=3 Hz, 2H), 8.44 (s, 1H), 8.49 (d, J=2.5 Hz, 1H), 10.21 (s, 1H), 13.83 (s, 1H); ESIMS found for C26H27N7O2 m/z 470.5 (M+H).




embedded image


5-(5-((3,3-Difluoropyrrolidin-1-yl)methyl)pyridin-3-yl)-N-(pyridin-3-yl)-1H-indazole-3-carboxamide 188.


White solid (209 mg, 0.48 mmol, 56.6% yield). 1H NMR (DMSO-d6) δ ppm 2.23-2.32 (m, 2H), 2.76 (t, J=7 Hz, 2H), 2.94 (t, J=13.5 Hz, 2H), 3.77 (s, 2H), 7.40 (q, J=8 Hz, 1H), 7.83 (dq, J=8 Hz, J=2 Hz, 2H), 8.04 (s, 1H), 8.31-8.34 (m, 2H), 8.49 (s, 1H), 8.53 (d, J=2 Hz, 1H), 8.85 (d, J=2.5 Hz, 1H), 9.08 (d, J=2 Hz, 1H), 10.70 (s, 1H), 14.01 (brs, 1H); ESIMS found for C23H20F2N6O m/z 435.2 (M+H).




embedded image


N-(Pyridin-3-yl)-5-(5-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 189.


White solid (30 mg, 0.08 mmol, 26.0% yield). 1H NMR (DMSO-d6) δ ppm 1.91-2.05 (m, 4H), 3.33-3.39 (m, 4H), 7.09 (s, 1H), 7.40 (q, J=8 Hz, 1H), 7.79 (s, 2H), 7.96 (d, J=2.5 Hz, 1H), 8.14 (s, 1H), 8.30-8.34 (m, 2H), 8.44 (s, 1H), 9.07 (d, J=2 Hz, 1H), 10.68 (s, 1H), 13.97 (brs, 1H); ESIMS found for C22H20N6O m/z 385.2 (M+H).




embedded image


5-(5-((Dimethylamino)methyl)pyridin-3-yl)-N-(6-(pyrrolidin-1-yl)pyridin-3-yl)-1H-indazole-3-carboxamide 190.


White solid (142 mg, 0.32 mmol, 39.7% yield). 1H NMR (DMSO-d6) δ ppm 1.92-1.97 (m, 4H), 2.20 (s, 6H), 3.35-3.40 (m, 4H), 3.53 (s, 2H), 6.46 (d, J=9 Hz, 1H), 7.80 (dq, J=9 Hz, J=1.5 Hz, 2H), 7.97 (dd, J=9 Hz, J=3 Hz, 1H), 8.00 (s, 1H), 8.46-8.50 (m, 3H), 8.82 (d, J=2.5 Hz, 1H), 10.22 (s, 1H), 13.86 (brs, 1H); ESIMS found for C25H27N7O m/z 442.4 (M+H).


Example 6

Preparation of N-(6-(2-fluorophenoxy)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide (18) is depicted below in Scheme 32.




embedded image



Step 1


To a solution of methyl 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylate (CXVII) (7.0 g, 20.6 mmol) in DMF (80 mL) and water (16 mL) was added K3PO4 (6.56 g, 30.9 mmol), pyridin-3-ylboronic acid (CXXXI) (2.79 g, 22.7 mmol), Pd(PPh3)4 (1.19 g, 1.03 mmol) and. The solution was purged with argon and heated at 90° C. for 3 h. The solution was cooled to room temperature and then concentrated under reduced pressure. The residue was dissolved in DCM and washed with water, dried over MgSO4, filtered and then evaporated under vacuum. The residue was purified on a silica gel column (100% DCM→1.5:98.5 MeOH:DCM) to give methyl 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylate (CXXXIX) as an orange oil which solidified at rt (6.28 g, 18.6 mmol, 90% yield). ESIMS found for C19H19N3O3 m/z 338.0 (M+H).


Step 2


Preparation of intermediate 5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxylic acid (CXL) was performed following the procedure listed in Scheme 25, Step 4. White solid (900 mg, 2.78 mmol, 15% yield). ESIMS found for C18H17N3O3 m/z 324.1 (M+H).


Step 3


Preparation of intermediate N-(6-(2-fluorophenoxy)pyridin-3-yl)-5-(pyridin-3-yl)-1-(tetrahydro-2H-pyran-2-yl)-1H-indazole-3-carboxamide (CXLI) was performed following the procedure listed in Scheme 28, Step 3. Off-white solid (207 mg, 0.41 mmol, 66% yield). 1H NMR (DMSO-d6) δ ppm 1.60-1.69 (m, 2H), 1.76-1.87 (m, 1H), 2.03-2.13 (m, 2H), 2.56-2.65 (m, 1H), 3.84 (dt, J=11 Hz, J=4 Hz, 1H), 3.99 (t, J=11 Hz, 1H), 6.07 (dd, J=10 Hz, J=2 Hz, 1H), 6.98 (dd, J=3 Hz, J=2 Hz, 1H), 7.03-7.08 (m, 2H), 7.14 (d, J=9 Hz, 1H), 7.46 (t, J=7 Hz, 1H), 7.61 (dd, J=8 Hz, J=5 Hz, 1H), 7.91 (dd, J=9 Hz, J=2 Hz, 1H), 8.05 (d, J=9 Hz, 1H), 8.25 (d, J=8 Hz, 1H), 8.37 (dd, J=9 Hz, J=3 Hz, 1H), 8.49 (s, 1H), 8.64 (dd, J=5 Hz, J=2 Hz, 1H), 8.66 (d, J=3 Hz, 1H), 9.00 (d, J=2 Hz, 1H), 10.59 (s, 1H); ESIMS found for C29H24FN5O3 m/z 509.2 (M+H).


Step 4


Preparation of N-(6-(2-fluorophenoxy)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide (18) was performed following the procedure listed in Scheme 28, Step 4. White solid (128 mg, 0.30 mmol, 54.7% yield). 1H NMR (DMSO-d6) δ ppm 7.16 (d, J=9 Hz, 1H), 7.23-7.39 (m, 4H), 7.52 (dd, J=8 Hz, J=5 Hz, 1H), 7.79-7.85 (m, 2H), 8.13 (td, J=8 Hz, J=2 Hz, 1H), 8.38 (dd, J=9 Hz, J=3 Hz, 1H), 8.46 (s, 1H), 8.56 (d, J=3 Hz, 1H), 8.59 (dd, J=5 Hz, J=1 Hz, 1H), 8.93 (d, J=2 Hz, 1H), 10.65 (s, 1H), 13.96 (brs, 1H); ESIMS found for C24H16FN5O2 m/z 426.0 (M+H).


The following compounds were prepared in accordance with the procedure described in the above Example 6.




embedded image


N-(6-(3-Fluorophenoxy)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 19.


Off-white solid (148 mg, 0.35 mmol, 89.3% yield). 1H NMR (DMSO-d6) δ ppm 6.98 (dd, J=8 Hz, J=2 Hz, 1H), 7.01-7.06 (m, 2H), 7.13 (d, J=9 Hz, 1H), 7.44 (q, J=7 Hz, 1H), 7.53 (dd, J=8 Hz, J=5 Hz, 1H), 7.80-7.85 (m, 2H), 8.14 (td, J=6 Hz, J=2 Hz, 1H), 8.40 (dd, J=9 Hz, J=3 Hz, 1H), 8.47 (s, 1H), 8.60 (dd, J=5 Hz, J=1 Hz, 1H), 8.69 (d, J=3 Hz, 1H), 8.93 (d, J=2 Hz, 1H), 10.71 (s, 1H), 13.99 (s, 1H); ESIMS found for C24H16FN5O2 m/z 426.0 (M+H).




embedded image


N-(6-(4-Fluorophenoxy)pyridin-3-yl)-5-(pyridin-3-yl)-1H-indazole-3-carboxamide 20.


White solid (82 mg, 0.19 mmol, 91.8% yield). 1H NMR (DMSO-d6) δ ppm 7.08 (d, J=9 Hz, 1H), 7.15-7.21 (m, 2H), 7.22-7.27 (m, 2H), 7.67 (dd, J=8 Hz, J=5 Hz, 1H), 7.81-7.88 (m, 2H), 8.31 (d, J=8 Hz, 1H), 8.36 (dd, J=9 Hz, J=3 Hz, 1H), 8.51 (s, 1H), 8.63 (d, J=3 Hz, 1H), 8.66 (dd, J=5 Hz, J=1 Hz, 1H), 9.02 (d, 2 Hz, 1H), 10.67 (s, 1H), 14.00 (s, 1H); ESIMS found for C24H16FN5O2 m/z 426.0 (M+H).


Example 7

Preparation of N-(6-carbamoylpyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (180) is depicted below in Scheme 33.




embedded image



Step 1


To a solution of N-(6-cyanopyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (62) (200 mg, 0.45 mmol) in glacial acetic acid (2 mL) heated at 85° C. was carefully added dropwise sulfuric acid (2 mL). The reaction was heated at 85° C. for another 20 minutes before pouring into ice. The solution was basified with cold 5N NH4OH. The solids formed were filtered, washed with cold washed and dried under vacuum. The dry solid was suspended in DCM and a few drops of MeOH were added. The insoluble solids were filtered and discarded. The filtrate was concentrated and suspended again in DCM, boiled for 15 minutes and filtered. The solid was dried under vacuum to give N-(6-carbamoylpyridin-3-yl)-5-(5-(piperidin-1-ylmethyl)pyridin-3-yl)-1H-indazole-3-carboxamide (180) as a white solid (192 mg, 0.42 mmol, 93.7% yield). 1H NMR (DMSO-d6) δ ppm 1.36-1.42 (m, 2H), 1.48-1.55 (m, 4H), 2.38 (brs, 4H), 3.56 (s, 2H), 7.49 (s, 1H), 7.65 (d, J=9 Hz, 1H), 7.80 (d, J=9 Hz, 1H), 7.97 (s, 1H), 8.03 (s, 2H), 8.41 (s, 1H), 8.45 (d, J=2 Hz, 1H), 8.54 (dd, J=9 Hz, J=2.5 Hz, 1H), 8.80 (d, J=2 Hz, 1H), 9.15 (d, J=2 Hz, 1H), 10.83 (brs, 1H); ESIMS found for C25H25N7O2 m/z 456.4 (M+H).


Administration and Pharmaceutical Compositions


Some embodiments include administration of the compounds described herein as pharmaceutical compositions comprising: (a) a safe and therapeutically effective amount of the indazole-3-carboxamide, or its corresponding enantiomer, diastereoisomer or tautomer, or pharmaceutically acceptable salt; and (b) a pharmaceutically acceptable carrier.


In some embodiments, the methods described herein further include administering the compounds of this invention in combination (administered together or sequentially) with other known agents.


Administration of the compounds disclosed herein or the pharmaceutically acceptable salts thereof can be via any of the accepted modes of administration for agents that serve similar utilities including, but not limited to, orally, subcutaneously, intravenously, intranasally, topically, transdermally, intraperitoneally, intramuscularly, intrapulmonarilly, vaginally, rectally, ontologically, neuro-otologically, intraocularly, subconjuctivally, via anterior eye chamber injection, intravitreally, intraperitoneally, intrathecally, intracystically, intrapleurally, via wound irrigation, intrabuccally, intra-abdominally, intra-articularly, intra-aurally, intrabronchially, intracapsularly, intrameningeally, via inhalation, via endotracheal or endobronchial instillation, via direct instillation into pulmonary cavities, intraspinally, intrasynovially, intrathoracically, via thoracostomy irrigation, epidurally, intratympanically, intracisternally, intravascularly, intraventricularly, intraosseously, via irrigation of infected bone, or via application as part of any admixture with a prosthetic devices. Oral and parenteral administrations are customary in treating the indications.


Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. Pharmaceutically acceptable compositions may include solid, semi-solid, liquid, solutions, colloidal, liposomes, emulsions, suspensions, complexes, coacervates and aerosols. Dosage forms, such as, e.g., tablets, capsules, powders, liquids, suspensions, suppositories, aerosols, implants, controlled release or the like. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, milling, grinding, supercritical fluid processing, coacervation, complex coacervation, encapsulation, emulsification, complexation, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose. The compounds can also be administered in sustained or controlled release dosage forms, including depot injections, osmotic pumps, pills (tablets and or capsules), transdermal (including electrotransport) patches, implants and the like, for prolonged and/or timed, pulsed administration at a predetermined rate.


The compounds can be administered either alone or more typically in combination with a conventional pharmaceutical carrier, excipient or the like. The term “excipient” is used herein to describe any ingredient other than the compound(s) of the invention. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-α-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, poloxamers or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, tris, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat. Cyclodextrins such as α-, β, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-b-cyclodextrins, or other solubilized derivatives can also be advantageously used to enhance delivery of compounds of the formulae described herein. Dosage forms or compositions containing a compound as described herein in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. The contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%, in a further embodiment 20-80%, Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 21st Edition (Lippincott Williams & Wilkins. 2005).


In one preferred embodiment, the compositions will take the form of a unit dosage form such as a pill or tablet and thus the composition may contain, along with the active ingredient, a diluent such as lactose, sucrose, dicalcium phosphate, or the like; a lubricant such as magnesium stearate or the like; and a binder such as starch, gum acacia, polyvinylpyrrolidine, gelatin, cellulose, cellulose derivatives or the like. In another solid dosage form, a powder, marume, solution or suspension (e.g., in propylene carbonate, vegetable oils, PEG's, poloxamer 124 or triglycerides) is encapsulated in a capsule (gelatin or cellulose base capsule). Unit dosage forms in which the two active ingredients are physically separated are also contemplated; e.g., capsules with granules (or tablets in a capsule) of each drug; two-layer tablets; two-compartment gel caps, etc. Enteric coated or delayed release oral dosage forms are also contemplated.


Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, etc. an active compound as defined above and optional pharmaceutical adjuvants in a carrier (e.g., water, saline, aqueous dextrose, glycerol, glycols, ethanol or the like) to form a solution, colloid, liposome, emulsion, complexes, coacervate or suspension. If desired, the pharmaceutical composition can also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, co-solvents, solubilizing agents, pH buffering agents and the like (e.g., sodium acetate, sodium citrate, cyclodextrin derivatives, sorbitan monolaurate, triethanolamine acetate, triethanolamine oleate, and the like).


In some embodiments, the unit dosage of compounds of Formula (I) is 0.25 mg/Kg to 50 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 0.25 mg/Kg to 20 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 0.50 mg/Kg to 19 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 0.75 mg/Kg to 18 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 1.0 mg/Kg to 17 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 1.25 mg/Kg to 16 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 1.50 mg/Kg to 15 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 1.75 mg/Kg to 14 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 2.0 mg/Kg to 13 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 3.0 mg/Kg to 12 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 4.0 mg/Kg to 11 mg/Kg in humans.


In some embodiments, the unit dosage of compounds of Formula (I) is 5.0 mg/Kg to 10 mg/Kg in humans.


In some embodiments, the compositions are provided in unit dosage forms suitable for single administration of a precise dose.


In some embodiments, the compositions are provided in unit dosage forms suitable for twice a day administration of a precise dose.


In some embodiments, the compositions are provided in unit dosage forms suitable for three times a day administration of a precise dose.


Injectables can be prepared in conventional forms, either as liquid solutions, colloid, liposomes, complexes, coacervate or suspensions, as emulsions, or in solid forms suitable for reconstitution in liquid prior to injection. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject. However, percentages of active ingredient of 0.01% to 10% in solution are employable, and could be higher if the composition is a solid or suspension, which could be subsequently diluted to the above percentages.


In some embodiments, the injection can be an intra-tendon injection.


In some embodiments, the composition will comprise 0.1-10% of the active agent in solution.


In some embodiments, the composition will comprise 0.1-5% of the active agent in solution.


In some embodiments, the composition will comprise 0.1-4% of the active agent in solution.


In some embodiments, the composition will comprise 0.15-3% of the active agent in solution.


In some embodiments, the composition will comprise 0.2-2% of the active agent in solution.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-96 hours.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-72 hours.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-48 hours.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-24 hours.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-12 hours.


In some embodiments, the compositions are provided in dosage forms suitable for continuous dosage by intravenous infusion over a period of 1-6 hours.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 5 mg/m2 to 300 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 5 mg/m2 to 200 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 5 mg/m2 to 100 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 10 mg/m2 to 50 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 50 mg/m2 to 200 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 75 mg/m2 to 175 mg/m2.


In some embodiments, these compositions can be administered by intravenous infusion to humans at doses of 100 mg/m2 to 150 mg/m2.


In one preferred embodiment, the compositions can be administered to the respiratory tract (including nasal and pulmonary) e.g., through a nebulizer, metered-dose inhalers, atomizer, mister, aerosol, dry powder inhaler, insufflator, liquid instillation or other suitable device or technique.


In some embodiments, aerosols intended for delivery to the nasal mucosa are provided for inhalation through the nose. For optimal delivery to the nasal cavities, inhaled particle sizes of about 5 to about 100 microns are useful, with particle sizes of about 10 to about 60 microns being preferred. For nasal delivery, a larger inhaled particle size is desired to maximize impaction on the nasal mucosa and to minimize or prevent pulmonary deposition of the administered formulation. In some embodiments, aerosols intended for delivery to the lung are provided for inhalation through the nose or the mouth. For optimal delivery to the lung, inhaled aerodynamic particle sizes of equal or less than 10 μm are useful, with an aerodynamic particle size of about 0.1 to 10 microns being preferred. Inhaled particles may be defined as liquid droplets containing dissolved drug, liquid droplets containing suspended drug particles (in cases where the drug is insoluble in the suspending medium), dry particles of pure drug substance, drug substance incorporated with excipients, liposomes, emulsions, colloidal systems, coacervates, aggregates of drug nanoparticles, or dry particles of a diluent which contain embedded drug nanoparticles.


In some embodiments, compounds of Formula (I) disclosed herein intended for respiratory delivery (either systemic or local) can be administered as aqueous formulations, as non-aqueous solutions or suspensions, as suspensions or solutions in halogenated hydrocarbon propellants with or without alcohol, as a colloidal system, as emulsions, coacervates or as dry powders. Aqueous formulations may be aerosolized by liquid nebulizers employing either hydraulic or ultrasonic atomization or by modified micropump systems (like the soft mist inhalers, the Aerodose® or the AERx® systems). Propellant-based systems may use suitable pressurized metered-dose inhalers (pMDIs). Dry powders may use dry powder inhaler devices (DPIs), which are capable of dispersing the drug substance effectively. A desired particle size and distribution may be obtained by choosing an appropriate device.


In some embodiments, the compositions of Formula (I) disclosed herein can be administered to the ear by various methods. For example, a round window catheter (e.g., U.S. Pat. Nos. 6,440,102 and 6,648,873) can be used.


Alternatively, formulations can be incorporated into a wick for use between the outer and middle ear (e.g., U.S. Pat. No. 6,120,484) or absorbed to collagen sponge or other solid support (e.g., U.S. Pat. No. 4,164,559).


If desired, formulations of the invention can be incorporated into a gel formulation (e.g., U.S. Pat. Nos. 4,474,752 and 6,911,211).


In some embodiments, compounds of Formula (I) disclosed herein intended for delivery to the ear can be administered via an implanted pump and delivery system through a needle directly into the middle or inner ear (cochlea) or through a cochlear implant stylet electrode channel or alternative prepared drug delivery channel such as but not limited to a needle through temporal bone into the cochlea.


Other options include delivery via a pump through a thin film coated onto a multichannel electrode or electrode with a specially imbedded drug delivery channel (pathways) carved into the thin film for this purpose. In other embodiments the acidic or basic solid gacyclidine can be delivered from the reservoir of an external or internal implanted pumping system.


Formulations of the invention also can be administered to the ear by intratympanic injection into the middle ear, inner ear, or cochlea (e.g., U.S. Pat. No. 6,377,849 and Ser. No. 11/337,815).


Intratympanic injection of therapeutic agents is the technique of injecting a therapeutic agent behind the tympanic membrane into the middle and/or inner ear. In one embodiment, the formulations described herein are administered directly onto the round window membrane via transtympanic injection. In another embodiment, the ion channel modulating agent auris-acceptable formulations described herein are administered onto the round window membrane via a non-transtympanic approach to the inner ear. In additional embodiments, the formulation described herein is administered onto the round window membrane via a surgical approach to the round window membrane comprising modification of the crista fenestrae cochleae.


In some embodiments, the compounds of Formula (I) are formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG (like PEG ointments), and the like. In suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.


Suppositories for rectal administration of the drug (either as a solution, colloid, suspension or a complex) can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt or erode/dissolve in the rectum and release the drug. Such materials include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, poloxamers, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.


In some embodiments, the compositions can be administered by transdermal patch.


Other modes of deliveries include using biodegradable or non-biodegradable scaffolds.


It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular patient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.


Solid compositions can be provided in various different types of dosage forms, depending on the physicochemical properties of the drug, the desired dissolution rate, cost considerations, and other criteria. In one of the embodiments, the solid composition is a single unit. This implies that one-unit dose of the drug is comprised in a single, physically shaped solid form or article. In other words, the solid composition is coherent, which is in contrast to a multiple unit dosage form, in which the units are incoherent.


Examples of single units which may be used as dosage forms for the solid composition include tablets, such as compressed tablets, film-like units, foil-like units, wafers, lyophilized matrix units, and the like. In a preferred embodiment, the solid composition is a highly porous lyophilized form. Such lyophilizates, sometimes also called wafers or lyophilized tablets, are particularly useful for their rapid disintegration, which also enables the rapid dissolution of the active compound.


On the other hand, for some applications the solid composition may also be formed as a multiple unit dosage form as defined above. Examples of multiple units are powders, granules, microparticles, pellets, mini-tablets, beads, lyophilized powders, and the like. In one embodiment, the solid composition is a lyophilized powder. Such a dispersed lyophilized system comprises a multitude of powder particles, and due to the lyophilization process used in the formation of the powder, each particle has an irregular, porous microstructure through which the powder is capable of absorbing water very rapidly, resulting in quick dissolution. Effervescent compositions are also contemplated to aid the quick dispersion and absorption of the compound.


Another type of multiparticulate system which is also capable of achieving rapid drug dissolution is that of powders, granules, or pellets from water-soluble excipients which are coated with the drug, so that the drug is located at the outer surface of the individual particles. In this type of system, the water-soluble low molecular weight excipient is useful for preparing the cores of such coated particles, which can be subsequently coated with a coating composition comprising the drug and, preferably, one or more additional excipients, such as a binder, a pore former, a saccharide, a sugar alcohol, a film-forming polymer, a plasticizer, or other excipients used in pharmaceutical coating compositions.


Also provided herein are kits. Typically, a kit includes one or more compounds or compositions as described herein. In certain embodiments, a kit can include one or more delivery systems, e.g., for delivering or administering a compound as provided above, and directions for use of the kit (e.g., instructions for treating a patient). In another embodiment, the kit can include a compound or composition as described herein and a label that indicates that the contents are to be administered to a patient with cancer. In another embodiment, the kit can include a compound or composition as described herein and a label that indicates that the contents are to be administered to a patient with one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease; and/or for promoting wound healing.


The actual dose of the active compounds of the present invention depends on the specific compound, and on the condition to be treated; the selection of the appropriate dose is well within the knowledge of the skilled artisan.


Methods of Treatment


The compounds and compositions provided herein can be used as inhibitors and/or modulators of one or more components of the Wnt pathway, which may include one or more Wnt proteins, and thus can be used to treat a variety of disorders and diseases in which aberrant Wnt signaling is implicated. Non-limiting examples include one or more diseases or conditions independently selected from the group consisting of a tendinopathy, dermatitis, psoriasis, morphea, ichthyosis, Raynaud's syndrome, and Darier's disease. In certain embodiments, the compounds and compositions provided herein can be used for promoting wound healing.


In some embodiments, the methods further include administering to a patient in need of such treatment an effective amount of one or more of the compounds of Formula (I), in combination (simultaneously or sequentially) with at least one other agent.


In some embodiments, the methods further include administering a pharmaceutical composition that includes a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient and optionally at least one other agent.


In some embodiments, the one or more diseases or conditions is a tendinopathy. In certain embodiments, the tendinopathy is tendinosis. In certain embodiments, the tendinopathy is tendinitis. In certain embodiments, the tendinopathy is tenosynovitis.


A tendon is a band of fibrous connective tissue that usually connects muscle to bone. Healthy tendons include parallel arrays of type I collagen fibers closely packed together, but also include a small amount of elastin and of proteoglycans. Tendons can be slow to heal if injured, and sometimes do not regain their original strength. Partial tears heal by the rapid production of disorganized type-III collagen, which is weaker than normal tendon. Recurrence of injury in the damaged region of tendon is common.


Tendons which may be treated by the methods of the invention include any tendon of the human or mammalian body. Non-limiting examples of tendons include the patellar tendon, the anterior tibialis tendon, the Achilles tendon, the hamstring tendon, the semitendinosus tendon, the gracilis tendon, the abductor tendon, the adductor tendon, the supraspinatus tendon, the infraspinatus tendon, the subscapularis tendon, the teres minor tendon, the flexor tendon, the rectus femoris tendon, the tibialis posterior tendon, and the quadriceps femoris tendon.


In some embodiments, the tendon is a tendon of the foot or ankle; e.g., the extensor hallucis longus, the flexor hallucis longus, the extensor digitorum longus, the extensor digitorum brevis, the peroneus longus, the peroneus brevis, the flexor hallucis brevis, the flexor digitorum longus, the posterior tibialis, the Achilles tendon, and the plantar fascia.


In some embodiments, the tendon is a tendon of the leg; e.g., the patellar tendon, the anterior tibialis tendon, the Achilles tendon, the hamstring tendon, the semitendinosus tendon, the gracilis tendon, the abductor tendon, the adductor tendon, the flexor tendon, the rectus femoris tendon, the tibialis posterior tendon, and the quadriceps femoris tendon.


In some embodiments, the tendon is a tendon of the shoulder; e.g., the supraspinatus tendon, the infraspinatus tendon, the subscapularis tendon, and the teres minor tendon (rotator cuff complex).


In some embodiments, the tendon is a tendon of the elbow; e.g., the biceps tendon, the triceps tendon, the extensor carpi radialis brevis, the common extensor tendon, the extensor digitorum, the extensor digiti minimi, the extensor carpi ulnaris, the supinator, the common flexor tendon, the pronator teres, the flexor carpi radialis, the palmaris longus, the flexor carpi ulnaris and the digitorum superficialis. In some embodiments, the tendon is a tendon of the wrist. In some embodiments, the tendon of the wrist is selected from the group consisting of biceps tendon, the triceps tendon, the extensor carpi radialis brevis, the common extensor tendon, the extensor digitorum, the extensor digiti minimi, the extensor carpi ulnaris, the supinator, the common flexor tendon, the pronator teres, the flexor carpi radialis, the palmaris longus, the flexor carpi ulnaris, the digitorum superficialis, the flexor pollicis brevis, the flexor pollicis longus, the abductor pollicis brevis, the abductor pollicis longus, the flexor digitorum profundus, the flexor digitorum superficialis, the extensor pollicis brevis, and the extensor pollicis longus. In some embodiments, the tendon is a tendon of the hand. In some embodiments, the tendon of the hand is selected from the group consisting of the flexor pollicis brevis, the flexor pollicis longus, the abductor pollicis brevis, the abductor pollicis longus, the flexor digitorum profundus, the flexor digitorum superficialis, the extensor pollicis brevis, and the extensor pollicis longus.


Non-limiting examples of tendinopathies include: clavicular or patellar tendinopathy, patellar tendonitis; medial tibial stress syndrome; Achilles tendinopathy, lateral epicondylitis or “tennis elbow;” medial epicondylitis or “golfer's elbow;” plantar fasciitis; and rotator cuff tendinopathy.


In some embodiments, the tendinopathy is rotator cuff tendinopathy; e.g., supraspinatus tendinopathy, infraspinatus tendinopathy, subscapularis tendinopathy, and teres minor tendinopathy.


In some embodiments, the tendinopathy is lateral epicondylitis or “tennis elbow” at the extensor muscle group origin at the lateral humeral condyle insertion, principally in the extensor carpi radialis brevis (ECRB) tendon. In some embodiments, the tendinopathy is medial epicondylitis or “golfer's elbow” at the interface between the pronator teres and flexor carpi radialis origin of the medial humeral condyle.


In some embodiments, the tendinopathy is patellar tendinopathy. In some embodiments, the tendinopathy is Achilles tendinopathy. In some embodiments, the tendinopathy is plantar fasciitis. In some embodiments, the tendinopathy is medial plantar fasciitis. In some embodiments, the tendinopathy is lateral plantar fasciitis.


In some embodiments, the tendinopathy is tendinosis. In some embodiments, the tendinosis is selected from the group consisting of extensor hallucis longus tendinosis, flexor hallucis longus tendinosis, extensor digitorum longus tendinosis, extensor digitorum brevis tendinosis, peroneus longus tendinosis, peroneus brevis tendinosis, flexor hallucis brevis tendinosis, flexor digitorum longus tendinosis, posterior tibialis tendinosis, Achilles tendon tendinosis, and plantar fascia tendinosis. In some embodiments, the tendinosis is selected from the group consisting of patellar tendinosis, the anterior tibialis tendinosis, the hamstring tendinosis, semitendinosus tendinosis, gracilis tendinosis, abductor tendinosis, and adductor tendinosis. In some embodiments, the tendinosis is selected from the group consisting of flexor tendinosis, rectus femoris tendinosis, tibialis posterior tendinosis, and quadriceps femoris tendinosis. In some embodiments, the tendinosis is selected from the group consisting of supraspinatus tendinosis, infraspinatus tendinosis, subscapularis tendinosis, and teres minor tendinosis.


In some embodiments, the tendinosis is selected from the group consisting of biceps tendinosis, triceps tendinosis, extensor carpi radialis brevis tendinosis, common extensor tendinosis, extensor digitorum tendinosis, extensor digiti minimi tendinosis, extensor carpi ulnaris tendinosis, supinator tendinosis, common flexor tendinosis, pronator teres tendinosis, flexor carpi radialis tendinosis, palmaris longus tendinosis, flexor carpi ulnaris tendinosis and digitorum superficialis tendinosis. In some embodiments, the tendinosis is selected from the group consisting of biceps tendinosis, triceps tendinosis, extensor carpi radialis brevis tendinosis, common extensor tendinosis, extensor digitorum tendinosis, extensor digiti minimi tendinosis, extensor carpi ulnaris tendinosis, supinator tendinosis, common flexor tendinosis, pronator teres tendinosis, flexor carpi radialis tendinosis, palmaris longus tendinosis, flexor carpi ulnaris tendinosis, digitorum superficialis tendinosis, flexor pollicis brevis tendinosis, flexor pollicis longus tendinosis, abductor pollicis brevis tendinosis, abductor pollicis longus tendinosis, flexor digitorum profundus tendinosis, flexor digitorum superficialis tendinosis, extensor pollicis brevis tendinosis, and extensor pollicis longus tendinosis. In some embodiments, the tendinosis is selected from the group consisting of flexor pollicis brevis tendinosis, flexor pollicis longus tendinosis, abductor pollicis brevis tendinosis, abductor pollicis longus tendinosis, flexor digitorum profundus tendinosis, flexor digitorum superficialis tendinosis, extensor pollicis brevis tendinosis, and extensor pollicis longus tendinosis.


In some embodiments, the tendinopathy is tendinitis. In some embodiments, the tendinitis is selected from the group consisting of extensor hallucis longus tendinitis, flexor hallucis longus tendinitis, extensor digitorum longus tendinitis, extensor digitorum brevis tendinitis, peroneus longus tendinitis, peroneus brevis tendinitis, flexor hallucis brevis tendinitis, flexor digitorum longus tendinitis, posterior tibialis tendinitis, Achilles tendon tendinitis, and plantar fascia tendinitis. In some embodiments, the tendinitis is selected from the group consisting of patellar tendinitis, the anterior tibialis tendinitis, the hamstring tendinitis, semitendinosus tendinitis, gracilis tendinitis, abductor tendinitis, and adductor tendinitis. In some embodiments, the tendinitis is selected from the group consisting of flexor tendinitis, rectus femoris tendinitis, tibialis posterior tendinitis, and quadriceps femoris tendinitis. In some embodiments, the tendinitis is selected from the group consisting of supraspinatus tendinitis, infraspinatus tendinitis, subscapularis tendinitis, and teres minor tendinitis.


In some embodiments, the tendinitis is selected from the group consisting of biceps tendinitis, triceps tendinitis, extensor carpi radialis brevis tendinitis, common extensor tendinitis, extensor digitorum tendinitis, extensor digiti minimi tendinitis, extensor carpi ulnaris tendinitis, supinator tendinitis, common flexor tendinitis, pronator teres tendinitis, flexor carpi radialis tendinitis, palmaris longus tendinitis, flexor carpi ulnaris tendinitis and digitorum superficialis tendinitis. In some embodiments, the tendinitis is selected from the group consisting of biceps tendinitis, triceps tendinitis, extensor carpi radialis brevis tendinitis, common extensor tendinitis, extensor digitorum tendinitis, extensor digiti minimi tendinitis, extensor carpi ulnaris tendinitis, supinator tendinitis, common flexor tendinitis, pronator teres tendinitis, flexor carpi radialis tendinitis, palmaris longus tendinitis, flexor carpi ulnaris tendinitis, digitorum superficialis tendinitis, flexor pollicis brevis tendinitis, flexor pollicis longus tendinitis, abductor pollicis brevis tendinitis, abductor pollicis longus tendinitis, flexor digitorum profundus tendinitis, flexor digitorum superficialis tendinitis, extensor pollicis brevis tendinitis, and extensor pollicis longus tendinitis. In some embodiments, the tendinitis is selected from the group consisting of flexor pollicis brevis tendinitis, flexor pollicis longus tendinitis, abductor pollicis brevis tendinitis, abductor pollicis longus tendinitis, flexor digitorum profundus tendinitis, flexor digitorum superficialis tendinitis, extensor pollicis brevis tendinitis, calcific tendinitis, and extensor pollicis longus tendinitis.


In some embodiments, the tendinitis is caused by chronic overuse injuries of tendon failed healing.


In some embodiments, the injury or damage is localized very near the muscle-tendon junction (myotendinous junction).


In some embodiments, the tendinitis leads to scarring and fibrosis.


The methods of the invention may result in improvement in one or more of the following: decreasing pain of the affected joint or limb, decreasing stiffness of the affected joint or limb, increasing mobility of the affected joint or limb, increasing strength of the affected joint or limb, decreasing the rate of tendinopathy progression, decreasing inflammation, increasing the strength of the tendon, or improving the rate of tendon strength recovery. Various methods for measuring effectiveness of the treatment include, but are not limited to: Disabilities of the Arm, Shoulder and Hand Score (DASH), Visual Analog Score (VAS), and grip strength testing.


In some embodiments, the treatment results in increased strength of the tendon. In some embodiments, the treatment results in a more rapid rate of tendon strength recovery. In some embodiments, the treatment results in an increase in tendon strength of about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90% within about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 days of administration of a compound of the invention, as compared to baseline.


The methods of the invention may include preventive treatments.


In some embodiments, the administering is by direct injection to the affected site. In some embodiments, the direct injection is accomplished using the “peppering technique” with or without ultrasound guidance. The “peppering technique” is an injection method whereby after the needle is inserted into the tender area, multiple small injections are performed by withdrawing, redirecting and reinserting the needle without emerging from the skin.


In some embodiments, the methods can further include administering one or more other therapeutic regimens and/or agents effective for treating a tendinopathy, e.g., palliative care, with treatment focusing on anti-inflammatory measures, including treatment with nonsteroidal anti-inflammatory drugs (NSAIDs), steroid injections, cortisone injections, platelet rich plasma (PRP) injections, physical therapy, shock wave therapy, low-level laser therapy (phototherapy), cell therapy, and sclerotherapy.


In some embodiments, the one or more diseases or conditions is psoriasis. Non-limiting examples include: psoriasis vulgaris (including nummular psoriasis and plaque psoriasis); generalized pustular psoriasis (including impetigo herpetiformis and von Zumbusch's disease); acrodermatitis continua; pustulosis palmaris et plantaris; guttate psoriasis; arthropathic psoriasis; other psoriasis (including inverse psoriasis).


In some embodiments, the one or more diseases or conditions is dermatitis. Non-limiting examples include: atopic dermatitis, contact dermatitis (e.g., allergic contact dermatitis, irritant contact dermatitis), stasis dermatitis, dermatitis that led up to steroid dermatitis, steroid-resistant dermatitis, dermatitis to which tacrolimus is not applicable, chronic dermatitis, erythroderma (e.g., erythroderma posteczematosa and erythroderma secondary to dermatoses, toxic erythroderma, infantile desquamative erythroderma, and paraneoplastic erythroderma), eczema, nummular eczema, dyshidrotic eczema, asteatotic eczema, seborrheic dermatitis, autosensitization dermatitis, stasis dermatitis, urticaria, drug eruption, dermal vasculitis, prurigo, pruritus cutaneus, erythema (e.g. nodosum or multiforme), rosacea, rosacea-like dermatitis, lichen planus, photo-induced dermatitis, or follicular keratosis. In certain embodiments, the dermatitis is contact dermatitis, e.g., allergic contact dermatitis, e.g., resulting from direct skin contact with a substance such as poison ivy, poison oak, or poison sumac.


In some embodiments, the one or more diseases or conditions is morphea.


In some embodiments, the one or more diseases or conditions is ichthyosis.


In some embodiments, the one or more diseases or conditions is Darier's disease.


In some embodiments, the methods can further include administering one or more other therapeutic regimens and/or agents effective in treating a skin disorder described herein, e.g., corticosteroids, immune modulators, vitamin D3 and its analogs, retinoic acids and their pharmaceutically active derivatives, or combinations thereof. Specific non-limiting examples of drugs include betamethasone dipropionate, clobetasol propionate, halobetasol propionate, diflorasone diacetate, amcinonide, desoximethasone, fluocinonide, halcinonide, mometasone furoate, betamethasone valerate, fluocinonide, fluticasone propionate, triamcinolone acetonide, fluocinolone acetonide, flurandrenolide, desonide, hydrocortisone butyrate, hydrocortisone valerate, alclometasone dipropionate, flumethasone pivolate, hydrocortisone, hydrocortisone acetate, prednisone, Benadryl, tacrolimus, picrolimus, tazarotene, isotretinoin, cyclosporin, anthralin, vitamin D3, cholecalciferol, calcitriol, calcipotriol, tacalcitol, calcipotriene, Gossypol, 4-hydroxyestradiol, 2-hydroxyestradiol, 2-hydroxyestrone, 2-benzimidazolylthioacetamide-N-ethyl-2-benzyl (KH7.102), an antibody, a nucleic acid, or combinations thereof.


In some embodiments, the one or more diseases or conditions is Raynaud's syndrome.


In some embodiments, the patient is a human.


In some embodiments, the compound of Formula (I) inhibits one or more proteins in the Wnt pathway.


In some embodiments, the compound of Formula (I) inhibits signaling induced by one or more Wnt proteins.


In some embodiments, the Wnt proteins are chosen from: WNT1, WNT2, WNT2B, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9B, WNT10A, WNT10B, WNT11, and WNT16.


In some embodiments, the compound of Formula (I) inhibits a kinase activity.


In some embodiments, the compound of Formula (I) inhibits one or more Wnt proteins.


Evaluation of Biological Activity


The biological activity of the compounds described herein can be tested using any suitable assay known to those of skill in the art, e.g., WO 2001/053268 or WO 2005/009997. For example, the activity of a compound may be tested using one or more of the test methods outlined below.


In another example, one may utilize in vitro assays for Wnt biological activity, e.g. stabilization of β-catenin and promoting growth of stem cells. Assays for biological activity of Wnt include stabilization of β-catenin, which can be measured, for example, by serial dilutions of a candidate inhibitor composition. An exemplary assay for Wnt biological activity contacts a Wnt composition in the presence of a candidate inhibitor with cells, e.g. mouse L cells. The cells are cultured for a period of time sufficient to stabilize β-catenin, usually at least about 1 hour, and lysed. The cell lysate is resolved by SDS PAGE, then transferred to nitrocellulose and probed with antibodies specific for β-catenin.


In a further example, the activity of a candidate compound can be measured in a Xenopus secondary axis bioassay (Leyns, L. et al. Cell (1997), 88(6), 747-756).


Example 8

Another screening assay for Wnt activity is described as follows. Reporter cell lines can be generated by stably transducing cells of cancer cell lines (e.g., colon cancer) with a lentiviral construct that include a Wnt-responsive promoter driving expression of the firefly luciferase gene.


Lentiviral constructs can be made in which the SP5 promoter, a promoter having eight TCF/LEF binding sites derived from the SP5 promoter, is linked upstream of the firefly luciferase gene. The lentiviral constructs can also include a hygromycin resistance gene as a selectable marker. The SP5 promoter construct can be used to transduce SW480 cells, a colon cancer cell line having a mutated APC gene that generates a truncated APC protein, leading to de-regulated accumulation of β-catenin. A control cell line can be generated using another lentiviral construct containing the luciferase gene under the control of the SV40 promoter which does not require β-catenin for activation.


Cultured SW480 cells bearing a reporter construct can be distributed at approximately 10,000 cells per well into 96 well or 384 well plates. Compounds from a small molecule compound library can then be added to the wells in half-log dilutions using a ten micromolar top concentration. A series of control wells for each cell type receive only buffer and compound solvent. Twenty-four to forty hours after the addition of compound, reporter activity for luciferase can be assayed, for example, by addition of the BrightGlo luminescence reagent (Promega) and the Victor3 plate reader (Perkin Elmer). Readings can be normalized to DMSO only treated cells, and normalized activities can then be used in the EC50 calculations. Table 2 shows the activity of selected indazole-3-carboxamide analogs.












TABLE 2





Compound
EC50 (nM)
Compound
EC50 (nM)




















1
175
nM
2
5,000
nM


3
200
nM
4
160
nM


5
10,000
nM
6
270
nM


7
110
nM
8
130
nM


9
10,000
nM
11
10,000
nM


12
63
nM
13
1,250
nM


14
106
nM
15
37
nM


16
10,000
nM
18
122
nM


19
107
nM
20
118
nM


23
120
nM
26
210
nM


32
1,250
nM
36
275
nM


37
1,120
nM
38
120
nM


39
65
nM
40
65
nM


41
67
nM
42
500
nM


43
63
nM
44
158
nM


45
110
nM
46
15
nM


47
71
nM
48
10,000
nM


49
57
nM
50
71
nM


51
26
nM
52
57
nM


53
63
nM
54
158
nM


55
44
nM
56
160
nM


57
10,000
nM
58
71
nM


59
3,100
nM
60
10,000
nM


61
239
nM
62
16
nM


63
100
nM
64
6
nM


65
101
nM
66
10,000
nM


67
10,000
nM
68
48
nM


69
50
nM
70
41
nM


71
25
nM
72
215
nM


73
322
nM
74
65
nM


75
40
nM
76
850
nM


77
2,650
nM
78
239
nM


79
123
nM
80
158
nM


81
77-142
nM
82
143-188
nM


83
2,500-3,400
nM
84
822-898
nM


86
66
nM
87
2,440
nM


106
33
nM
124
67
nM


126
22
nM
162
426
nM


163
15,400
nM
168
66
nM


169
49
nM
170
43
nM


172
60
nM
173
36
nM


174
48
nM
175
25
nM


176
30
nM
177
183
nM


178
297
nM
179
30
nM


180
13
nM
181
38
nM


182
35
nM
183
49
nM


184
40
nM
185
27
nM


186
460
nM
187
215
nM


188
9
nM
189
85
nM


190
1,200
nM









Example 9

Representative compounds were screened using primary human mesenchymal stem cells (hMSCs) to determine their ability to induce tenocyte differentiation (process by which tendon is developed).


Human Mesenchymal Stem Cell Culture:


Primary human mesenchymal stem cells (hMSCs) were purchased from Lonza (Walkersville, Md.) and expanded in Mesenchymal Stem Cell Growth Media (Lonza). Cells between passage 3 and 6 were used for the experiments.


Compound Screening:


Each compound was dissolved in DMSO as a 10 mM stock and used to prepare compound source plates. For the tenocyte differentiation assay, serial dilution (1:2, 10-point dose-response curves from 10 μM to 19.5 nM) and compound transfer was performed using the ECHO 550 (Labcyte, Sunnyvale, Calif.) into 384-well black clear bottom assay plates (Greiner Bio-One) with appropriate DMSO backfill for a final DMSO concentration of 0.17%. hMSCs were plated at 3,000 cells/well in 70 μL/well Dulbecco's modified Eagle's medium (DMEM, Life Technologies, Carlsbad, Calif.) with 1% Fetal Bovine Serum (FBS, Life Technologies). Bone Morphogenic Factor (BMP) and Fetal Growth Factor (FGF) (10 ng/ml each, Peprotech, Inc., Rocky Hill, N.J.) were used as a positive-controls for differentiation while negative control wells were treated with 120 μL DMSO for normalization and calculating EC50 values. Cells were incubated at 37° C. and 5% CO2 for 4 days. Cells were fixed using 4% formaldehyde (Electron Microscopy Sciences), and stained with anti-Sclerasis (anti-SCXA) antibodies (Abgent, San Diego, Calif.) [Webb S., et. al., Retinoic acid receptor signaling preserves tendon stem cell characteristics and prevents spontaneous differentiation in vitro, Stem Cell Research & Therapy 2016, 7:45] overnight at 4° C. The cells were washed with Phosphate Buffered Saline (PBS, Life Technologies) and incubated with anti-rabbit Alexa-flor 647 secondary antibodies (Life Technologies) and DAPI (Life Technologies) for 1 hour at room temperature. Cells were washed using PBS, and imaged using the CellInsight CX5 (Life Technologies, 594/633 nm filter). Number of cells positive for SCXA in each well was quantified using the Celllnsight CX5. Data was normalized to the average of 12 DMSO treated wells on the same plate using Dotmatics Studies module. The normalized averages (fold change over DMSO) of 3 replicate wells for each compound concentration were calculated. Due to solubility limitations of some of the compounds, values for higher doses were manually corrected and curve fitting and EC50 determinations were performed using Dotmatics Studies.


EC50 for each compound is reported. Table 3 shows the measured activity for representative compounds of Formula I as described herein.












TABLE 3







Compound
EC50 (μM)



















1
>100 uM



2
>100 uM



3
>100 uM



4
4.552



5
>100 uM



6
92.468 



7
6.942



8
4.983



9
>100 uM



11
>100 uM



12
4.998



13
>100 uM



14
50.395 



15
4.804



16
35.772 



18
5.378



19
>100 uM



20
11.982 



23
>100 uM



26
>100 uM



32
>100 uM



36
>100 uM



37
>100 uM



38
2.839



39
>100 uM



40
5.174



41
8.118



42
5.137



43
5.019



44
25.061 



45
9.750



46
5.056



47
>100 uM



48
>100 uM



49
>100 uM



50
>100 uM



51
>100 uM



52
>100 uM



53
>100 uM



54
>100 uM



55
>100 uM



56
0.625



57
>100 uM



58
>100 uM



59
14.560 



60
>100 uM



61
12.268 



62
>100 uM



63
10.243 



64
7.661



65
>100 uM



66
>100 uM



67
>100 uM



68
4.613



69
12.481 



70
10.850 



71
>100 uM



72
>100 uM



73
>100 uM



74
10.180 



75
12.373 



76
2.667



77
6.075



78
>100 uM



79
>100 uM



80
>100 uM



81
>100 uM



82
>100 uM



83
>100 uM



84
>100 uM



86
5.273



87
1.584



106
5.545



124
>100 uM



126
5.537



162
>100 uM



163
5.086



168
>100 uM



169
4.905



170
>100 uM



172
>100 uM



173
>100 uM



174
5.539



175
10.377 



176
4.654



177
5.174



178
>100 uM



179
1.115



180
>100 uM



181
>100 uM



182
8.039



183
>100 uM



184
>100 uM



185
6.442



186
>100 uM



187
>100 uM



188
1.212



189
>100 uM



190
>100 uM










Example 10

Wnt Signaling inhibition


Cell Culture.


SW480 cells (ATCC) were cultured in Dulbecco's modified Eagle's medium (DMEM, ThermoFisher) with 1% Glutamax (Life Technologies), 1% Penicillin-Streptomycin (Life Technologies), 150 ug/ml Hygromycin (Life Technologies) with 10% fetal bovine serum (FBS) (Hyclone) at 37° C., 5% CO2.


Wnt Reporter Assay. Human colorectal cancer cell line SW480, stably expressing the Wnt responsive promoter linked to luciferase gene, was plated overnight at 4×10e4 cells/well in DMEM, high glucose, no glutamine, no phenol red (Life Technologies), 1% Glutamax (Life Technologies), 1% Sodium Pyruvate (Life Technologies), 1% Penicillin-Streptomycin (Life Technologies) with 1% Fetal Bovine Serum (Hyclone). Cells were then treated with DMSO (vehicle control) or COMPOUND 175 at 10 μM top concentration and half a log dilution up to 10 concentrations (10, 3, 1, 0.3, 0.1, 0.03, 0.01, 0.003, 0.001 and 0.0003 μM). Cells were incubated for 48 hours at 37° C. 15 μl of Bright-Glo (Bright-Glo™ Luciferase Assay System, Promega) was added to the cells and luminescence was measured using the Cytation 3 plate reader (Biotek). Data was normalized using the DMSO control and inhibition profile and EC50 was calculated using Prism 4 (GraphPad Software Inc, La Jolla, Calif., USA). Prism 4.0 was used to calculate the EC50 of the Wnt reporter inhibition assay.


Exposure of the SW480 cells to compound 175 at concentrations of 10, 3, 1, 0.3, 0.1, 0.03, 0.01, 0.003, 0.001 and 0.0003 μM showed that compound 175 inhibited Wnt pathway activity in these cells in a dose-dependent manner with EC50 of 152.9 nM (see FIG. 1).


Example 11

Tendon Differentiation


Cell Culture.


Primary human mesenchymal stem cells (hMSCs; Lonza Inc) were cultured in MSCGM™ Mesenchymal Stem Cell Growth Medium, with 10% FBS (Lonza) 10% fetal bovine serum (FBS) (Hyclone) at 37° C., 5% CO2. hMSCs were used between passage 2 and 6 and were never allowed to reach confluence to maintain a naive state.


Tenocyte Differentiation Assay. For tenocyte differentiation, hMSCs were plated in 384-well plates at 1×104 cells/well in Dulbecco's modified Eagle's medium (DMEM, ThermoFisher) with 1% fetal bovine serum (FBS) (Hyclone). Cells were then treated with DMSO (vehicle control) or compound 175 at 750 nM top concentration and 2-fold dilution up to 8 concentrations (750, 333.3, 166.6, 83.3, 41.7, 21.7, 10.8 and 5.8 nM) and incubated for 7 days at 37° C. A combination of BMP and FGF (long/ml each, Peprotech, Inc.) was used as a positive control. At the end of 7 days, cells were fixed using 4% methanol-free formaldehyde (Electron Microscopy Sciences) for 10 min, washed with phosphate buffered saline (PBS) 3 times, permeabilized with PBS containing 0.3% triton X-100 (Sigma) for 5 min, blocked with PBST (PBS containing 0.3% triton X-100) with 3% bovine serum albumin (BSA; Sigma) for 1 h at room temperature, followed by incubation with primary antibodies in PBST+BSA overnight at 4° C. Cells were rinsed 3 times with PBS and incubated with fluorophore-conjugated secondary antibody in PBST+BSA and DAPI for 1 h at room temperature and washed with PBS 3 times. Plates were imaged using a CX5 high-content imager (ThermoFisher) and the % of cells stained positive were determined using a cell scoring algorithm (ThermoFisher). Data was plotted and EC50 was calculated using Prism 5 (GraphPad Software Inc, La Jolla, Calif., USA). Prism 5.0 was used to calculate the EC50 of the Tenocyte differentiation assay.


Exposure of the hMSCs to compound 175 at concentrations of 750, 333.3, 166.6, 83.3, 41.7, 21.7, 10.8 and 5.8 nM showed that COMPOUND 175 induced the expression of SCXA, TenacinC and Tenomodulin, in a dose-dependent manner with an EC50 between 139-189 nM (see FIGS. 2A-C and 3), indicating a differentiation of hMSC into tenocytes.


Anti-Inflammation


Cell Culture.


THP-1 cells (Catalog # TIB-202, ATCC, Manassas, Va.) were cultured in Roswell Park Memorial Institute (RPMI) 1640 Medium (Catalog #21870-100, Buffalo, N.Y.) with 1% L-glutamine, 1% HEPES, 1% Sodium Pyruvate, 2% Sodium Bicarbonate supplemented with 100 units/mL penicillin, 50 μg/mL streptomycin, 2-mercaptoethanol (0.05 mM) [basal medium] and 10% fetal bovine serum (Catalog #16140089, Life Technologies, Carlsbad, Calif.) at 37° C. and 5% CO2.


Cytokine Production Assay.


THP-1 cells were cultured in basal media with 1% FBS for 24 hours before the start of the assay. THP-1 cells were plated at 6×10e4 cells/well and treated with DMSO (vehicle control) or compound 175 at 10 μM highest test concentration and 2 to 3.5-fold serial dilutions up to 8 concentrations (10, 3.5, 1, 0.5, 0.1, 0.035, 0.01, 0.005 μM). For the TNFα assay, 50 ng/mL of LPS was added to the wells after 2 hours to induce cytokine production, and cells were incubated for 5 hours at 37° C. For the IL6 assay, 500 ng/ml of LPS was added after 2 hours and cells were incubated for 22 hours at 37° C. Plates were spun in a centrifuge for 1 minute at 10,000 rpm and supernatants were collected for ELISA. Supernatants were diluted 1:1 for the TNFα assay and 1:4 for the IL6 assay using the assay medium. ELISA was performed using Human TNF-α ELISA MAX™ Deluxe (Catalog #430204, Biolegend, San Diego, Calif.) and Human IL-6 ELISA MAX™ Deluxe (Catalog #430504, Biolegend, San Diego, Calif.) kits. Briefly, 96-well plates were coated with the appropriate capture antibody overnight and washed to remove excess antibody. Blocking buffer was added and incubated for 1 hour to prevent non-specific binding. Diluted supernatants were incubated in the coated plates for 2 hours at room temperature. Following washes to remove unbound proteins, biotinylated detection antibody was added and incubated for 30 mins at room temperature, followed by washes to remove unbound excess antibody. Avidin-HRP was then added and incubated for 30 mins at room temperature. Following several washes to remove unbound avidin-HRP, the TMB substrate was added and the plates were read on the Cytation 3 plate reader (Biotek Inc., Winooski, Vt.) at an absorbance of 450 nm with correction at 570 nm. All samples were processed in triplicate. Inhibition profile and EC50 was calculated using Prism 5 (GraphPad Software Inc, La Jolla, Calif., USA). Prism 5.0 was used to calculate the EC50 of the TNFα and IL6 inhibition assays.


Exposure of THP-1 cells to LPS induced the production of both TNFα and IL6. Compound 175 treatment inhibited the LPS-induced cytokine production in these cells in a dose-dependent manner with average EC50 ranging from 342-547 nM for TNFα and 356-629 nM for IL6 (see FIGS. 4A-B), with results generated from two independent assays and three replicates per assay.


Example 12

Rat Tendinopathy Model (1)


Rat Model of Collagenase-Induced Tendonitis.


Fifty (50 μl) of Collagenase Type IA (10 mg/ml in PBS, pH 7.4, approximately 469 units/mg) (Catalog # C5138, Sigma, St. Louis, Mo.) was injected into the Achilles tendon of both ankles near the osteotendinous junction of male Sprague Dawley CD® IGS rats (CRL, Inc.), using insulin syringes with 28.5 G needle.


Delivery of Compound 175 by Topical Application.


Rats were divided into 6 groups with 5 animals per group: Group 1 (sham injection); Group 2 (Collagenase-Vehicle); Group 3 (Collagenase-compound 175 10 mg/mL with 1% BA) and Group 4 (Collagenase-COMPOUND 175 10 mg/mL with 0.5% Tween 80). COMPOUND 175, formulated in 1% HPMC 40-0101/20% PG/1% BA or 1% HPMC 40-0101/20% PG/0.5% Tween 80, or vehicle (1% HPMC 40-0101/20% PG/1% BA) was applied at a dose volume of 30 μL/cm2 to cover a 2 cm2 dose area to the skin near the Achilles tendon region of both ankles of the collagenase-injected rats using an applicator and rubbed into the skin for 10 seconds. All rats were periodically observed for any pain, illnesses or abnormalities. At various time points, blood was collected for plasma in lithium heparin coated tubes by saphenous vein bleeding. For histology, rats were euthanized using isoflurane, tendons were isolated (n=6 tendons/group), fixed in 10% buffered formalin (Catalog # SF93-4, Fisher, Pittsburgh Pa.) and H & E histology staining was performed at a secondary site (Pacific Pathology, San Diego, Calif.). For biochemical analysis, tendons were isolated (n=4 tendons/group), flash frozen in liquid nitrogen and then stored at −80° C. for subsequent biochemical analysis (not reported).


Histology Scoring of Tendons.


There is biological variability in response to collagenase when injected directly into the tendons. This results in varying degrees of severity and location of inflammation as well as tendon degeneration (Urdzikova et al., Human multipotent mesenchymal stem cells improve healing after collagenase tendon injury in the rat. Biomed Eng Online. 2014 Apr. 9; 13:42). Therefore, the entire tendon was isolated and placed in a tissue cassette and embedded in paraffin. Five (5) μm sections were generated and an average of 3 sections were fixed on one slide, with a minimum of eight (8) slides per tendon and subjected to Hematoxylin & Eosin (H&E) staining. Slides were viewed under an upright light microscope (EVOS AMEX-1200) at a 10× magnification and scored blinded using a 1-4 scoring system of severity: 1—representing severe damage and 4—representing normal tendon, for Linearity of the fiber structure, Shape of the tendon cells, Density of the tendon cells, Inflammation, and Hemorrhage for a total score of twenty (20) using a modification of that described previously (Urdzikova et al., 2014—see Table 3). After the analysis was completed, the scorer was unblinded and scores for each criterion was annotated, totaled and total scores from each section were averaged together to generate a final score for each rat followed by averaging of scores for a given treatment group.


Biomarker Assays in Plasma.


Following manufacturer's instructions, KC/GRO plasma levels were determined using the KC/GRO Immunoassay kit (Catalog#900-K57, Peprotech, Rocky Hill, N.J.). Briefly, 96-well plates were coated with the anti-rat KC/GRO capture antibody overnight and washed to remove excess antibody. Plasma samples were incubated in the coated plates for 2 hours at room temperature. Following washes to remove unbound proteins, biotinylated anti-rat KC/GRO detection antibody was added and incubated for 30 mins at room temperature. Following washes to remove unbound excess antibody, avidin-HRP was added and incubated for 30 mins at room temperature. Following washes to remove unbound Avidin-HRP, the ABTS substrate was added and the plates were read on the Cytation 3 plate reader (Biotek Inc., Winooski, Vt.) at an absorbance of 525 nm with correction at 450 nm. All samples were processed in triplicate.


Data Analysis.


For tendon scoring, each section was analyzed following a 1-4 point scoring system as previously described (Urdzikova et al., 2014) with the exclusion of one variable, the Thickness of the epitenon, due to the difficulty in discerning the epitenon layer in histology sections. Table 4 below briefly summarizes the scoring system.












TABLE 4







Variablea
Score and Criteria









Linearity of the fiber structure
1 = no linear areas




2 = 20-50% linear




3 = >50% linear




4 = linear (normal)



Shape of the tendon cells
1 = predominantly round




2 = moderately round




3 = slightly oval




4 = linear (normal)



Density of the tendon cells
1 = sheets of cells




2 = moderate increase




3 = slight increase




4 = sparse(normal)



Inflammation
1 = severe increase




2 = moderate increase




3 = slight increase




4 = none



Hemorrhage
1 = predominant hemorrhage




2 = multiply areas in each field




3 = sparse or patchy




4 = none








aHistology Scoring was modified by Samumed to exclude one variable, Thickness of the epitenon.







In this study, a total of 64 sections were scored for Group 1 (sham injection group) and approximately 96 sections each for Group 2 (Collagenase-Vehicle), Group 3 (Collagenase-compound 175 with BA preservative) group and Group 4 (Collagenase-compound 175 without preservative). To determine statistical significance between groups, student's t-test was performed with p<0.05 considered as significant for histology scoring and biomarker assay.


Compound 175 Ameliorates Collagenase-Induced Tendinopathy.


Collagenase induced Tendonitis in rats recapitulates acute tendon injury in humans with inflammation resulting within a few hours (Urdzikova et al., 2014). Using an interventional approach, topical formulation of compound 175 was administered 24 hours post collagenase injection into the Achilles tendon. The Collagenase-injected rats were dosed with vehicle alone or compound 175 (10 mg/mL) formulated with or without BA as a preservative for 21 consecutive days. Group 1 rats that received sham injections served as a control for no disease induction.


As shown in FIG. 5, a significant presence of inflammatory cells was evident in the Collagenase-vehicle group. Consequently, there was evidence of tendon degeneration and damage in the Collagenase-vehicle group compared to normal tendons found in the sham control group. Amelioration of inflammation as well as tendon degeneration by compound 175 is demonstrated in FIG. 5. The histopathology of the tendon from the treatment groups revealed tendons with decreased inflammation as well as improved structure of the fibers and tendon cells with respect to linearity, shape and density. This observation was further confirmed by blinded Histology scoring. As shown in FIG. 6, upon topical treatment with compound 175, both compound 175 treatment groups demonstrated statistically significant increases in the tendon scores, achieving a score of 14.0 (±0.217) in Group 3 (Collagenase-compound 175 with BA) and a score of 12.2 (±0.284) in Group 4 (Collagenase-compound 175 without BA), compared to a score of 9.14 (±2.43) in Group 2 (Collagenase-vehicle), with a p-value of <0.01 and <0.05, respectively by student's t test.


Compound 175 Reduces Plasma Biomarker KC/GRO in the Collagenase-Induced Tendon Injury Model.


KC/GRO is an inflammatory biomarker reported to be associated with the development of tendonitis. In this study, plasma concentrations of KC/GRO were investigated at various timepoints of the study. As shown in Table 4 and FIG. 7, KC/GRO levels were elevated in the Group 2 (Collagenase-vehicle) on Days 5-21, ranging from 814-1483 pg/mL, while both the COMPOUND 175-treated groups, with or without BA, had lower levels of KC/GRO in plasma, ranging from 400-480 pg/mL and 347-451 pg/mL, respectively. Both compound 175-treated groups (Groups 3 and 4) demonstrated a statistically significant decrease in plasma KC/GRO levels compared to Collagenase-vehicle control on Days 7 and 21 (p<0.05 by student's t test, Table 5). Overall, the decrease of a plasma biomarker of inflammation corroborates the ability of compound 175 to ameliorate tendonitis.











TABLE 5









Plasma KC/GRO Concentration (pg/mL)












Group 3
Group 4



Group 2
Compound 175
Compound 175



Vehicle
(10 mg/mL with
(10 mg/mLwithout BA,


Day
(0 mg/mL)
0.5% BA)
with 0.5% Tween 80)













0
 228 ± 57.4
332 ± 51.3
530 ± 31.4


1
574 ± 106
521 ± 23.9
637 ± 120 


3
376 ± 156
813 ± 61.4
541 ± 58.9


5

1185 ± 4851)

480 ± 48.7
410 ± 86.0


7
1483 ± 286a 
 421 ± 57.2*
 451 ± 76.6*


14
1356 ± 367a 
400 ± 10.3
424 ± 76.2


21
814 ± 92.4a
 408 ± 83.3*
 347 ± 111*






aMean values were above quantitation limit (AQL > 1000 pg/mL) and are reported as estimated.



*p < 0.05 (student's t test) in Compound 175-treated groups (Groups 3 and 4) as compared to Group 2 (Vehicle) on Days 7 and 21.






Tendinopathy is an acute injury of the tendon that involves inflammation and tendon damage. If left untreated, repeated injury can lead to tendon ruptures and require surgery. The presence or absence of the preservative benzyl alcohol has no effect on the efficacy of compound 175. Treatment with compound 175 ameliorates tendinopathy as assessed by blinded histological scoring of the tendon. To further confirm this finding, it has shown that compound 175 also results in a decrease of an inflammatory plasma biomarker, KC/GRO.


Example 13

Rat Tendinopathy Model (2)


Fifty (50 μl) of Collagenase Type IA (10 mg/ml in PBS, pH 7.4, approximately 469 units/mg) (Catalog # C5138, Sigma, St. Louis, Mo.) was injected into the Achilles tendon of both ankles near the osteotendinous junction of male Sprague Dawley CD® IGS rats (CRL, Inc.), using insulin syringes with 28.5 G needle.


Delivery of Compound 175 by Topical Application.


Rats were divided into 6 groups, with 6 animals per group except 8 animals in Group 2: Group 1 (sham injection), Group 2 (Collagenase-Vehicle with 0.5% Phx), Group 3 (Collagenase-compound 175 3 mg/mL with 0.5% BA), Group 4 (Collagenase-compound 175 10 mg/mL with 0.5% BA), Group 5 (Collagenase-compound 175 3 mg/mL with 0.5% Phx) and Group 6 (Collagenase-compound 175 10 mg/mL with 0.5% Phx). COMPOUND 175, either at 3 or 10 mg/ml, prepared in either 1% HPMC 40-0101/20% PG/0.5% BA or 1% HPMC 40-0101/20% PG/0.5% Phx or vehicle (1% HPMC 40-0101/20% PG/0.5% Phx) was applied at 30 μL/cm2 over a 2 cm2 area to the skin near the Achilles tendon region of both ankles of the collagenase-injected rats using an applicator, and rubbed into the skin for 10 seconds. All rats were periodically observed for any pain, illnesses or abnormalities. At various time points, blood was collected for plasma in lithium heparin coated tubes by saphenous vein bleeding. For histology, rats were euthanized using isoflurane, tendons were isolated (n=8 tendons/group), fixed in 10% buffered formalin (Catalog # SF93-4, Fisher, Pittsburgh, Pa.) and H & E histology staining was performed at a secondary site (Pacific Pathology, San Diego, Calif.). For biochemical analysis, tendons were isolated (n=4 tendons/group), flash frozen in liquid nitrogen and then stored at −80° C. for subsequent biochemical analysis (not reported). Additionally, one (1) day after collagenase injection, 4 tendons were collected from Group 2 (vehicle) to observe the induction of inflammation (not reported).


There is biological variability in response to collagenase when injected directly into the tendons. This results in varying degrees of severity and location of inflammation as well as tendon degeneration (Urdzikova et al., 2014). Therefore, the entire tendon was isolated and placed in a tissue cassette and embedded in paraffin. Five (5) μm sections were generated with an average of 3 sections being fixed on one slide with a minimum of eight (8) slides per tendon and subjected to Hematoxylin & Eosin (H&E) staining. Slides were viewed under an upright light microscope (EVOS AMEX-1200) at a 10× magnification and scored blinded using a 1-4 scoring system of severity: 1-representing severe damage and 4—representing normal tendon, for Linearity of the fiber structure, Shape of the tendon cells, Density of the tendon cells, Inflammation, and Hemorrhage for a total score of twenty (20) using a modification of that described previously (Urdzikova et al., 2014—see Table 3). After the analysis was completed, the scorer was unblinded and scores for each criterion was annotated, totaled and total scores from each section were averaged together to generate a final score for each rat followed by averaging of scores for a given treatment group.


Biomarker Assays in Plasma.


Following manufacturer's instructions, KC/GRO plasma levels were determined using the KC/GRO Immunoassay kit (Cat #900-K57, Peprotech, Rocky Hill, N.J.). Briefly, 96-well plates were coated with the anti-rat KC/GRO capture antibody overnight and washed to remove excess antibody. Plasma samples were incubated in the coated plates for 2 hours at room temperature. Following washes to remove unbound proteins, biotinylated anti-rat KC/GRO detection antibody was added and incubated for 30 mins at room temperature. Following washes to remove unbound excess antibody, avidin-HRP was added and incubated for 30 mins at room temperature. Following washes to remove unbound avidin-HRP, the ABTS substrate was added and the plates were read on the Cytation 3 plate reader (Biotek Inc., Winooski, Vt.) at an absorbance of 525 nm with correction at 450 nm. All samples were processed in triplicate.


Data Analysis.


For tendon scoring, each section was analyzed following a 1-4 point scoring system as previously described (Urdzikova et al., 2014) with the exclusion of one variable, Thickness of the epitenon, due to difficulty in discerning the epitenon layer in the histology sections. Table 2 below briefly summarizes the scoring system. In this study, a total of 94 sections were scored for Group 1 (sham injection, control group) and between 92-116 sections for Group 2 (Collagenase-Vehicle with 0.5% Phx); Group 3 (Collagenase-compound 175 3 mg/mL with 0.5% BA), Group 4 (Collagenase-compound 175 10 mg/mL with 0.5% BA), Group 5 (Collagenase-compound 175 3 mg/mL with 0.5% Phx) and Group 6 (Collagenase-compound 175 10 mg/mL with 0.5% Phx).


Compound 175 Ameliorates Collagenase-Induced Tendinopathy.


Collagenase-induced tendonitis in rats recapitulates acute tendon injury in humans with inflammation resulting within a few hours (Urdzikova et al., 2014). Using an interventional approach, topical formulation of compound 175 was administered one day (˜24 hours) after the injection of collagenase into the Achilles tendon. The collagenase-injected rats were dosed with vehicle alone (containing 0.5% Phx) or compound 175 (3 or 10 mg/mL) formulated with either 0.5% BA or 0.5% Phx as a preservative for twenty-one (21) consecutive days. Group 1 rats (sham injections) served as control animals for no disease induction.


As shown in FIG. 8, a significant presence of inflammatory cells was evident in the Collagenase-vehicle group (Group 2). Consequently, there was evidence of tendon degeneration and damage in the Collagenase-vehicle group compared to normal tendons found in the sham control group (Group 1). Amelioration of inflammation as well as tendon degeneration by compound 175 is demonstrated in FIG. 8. The histopathology of the tendon from the compound 175-treated groups revealed tendons with decreased inflammation as well as improved structure of the fibers and tendon cells with respect to linearity, shape and density. This observation was further confirmed by blinded Histology scoring. As shown in FIG. 9, upon topical treatment with compound 175, all four (4) compound 175-treated groups demonstrated statistically significant increases in the tendon scores, achieving a score of 14.5 (±1.15) in Group 3 (Collagenase-compound 175 3 mg/mL with 0.5% BA), a score of 14.8 (±0.968) in Group 4 (Collagenase-compound 175 10 mg/mL with 0.5% BA), a score of 15.1 (±0.506) in Group 5 (Collagenase-compound 175 3 mg/mL with 0.5% Phx) and a score of 13.2 (±0.855) in Group 6 (Collagenase-compound 175 10 mg/mL with 0.5% Phx), compared to a score of 8.57 (±0.672) in Group 2 (Collagenase-vehicle), with p-values of <0.001 (Groups 3, 4 and 5) and <0.01 (Group 6) versus Group 2, by student's t test.


Compound 175 Reduces Plasma Biomarker KC/GRO in the Collagenase-Induced Tendon Injury Model.


KC/GRO is an inflammatory biomarker reported to be associated with the development of tendonitis. In this study, plasma concentrations of KC/GRO were investigated at various timepoints of the study. As shown in Table 5 and FIG. 10, KC/GRO levels were elevated in Group 2 (Collagenase-vehicle with 0.5% Phx) on Days 5-21, ranging from 488-778 pg/mL, while all four compound 175-treated groups had decreased levels of KC/GRO in plasma, ranging from 150-561 pg/mL. Compound 175 at both 3 and 10 mg/mL dose concentrations, with either BA or Phx as preservative (Groups 3-6), demonstrated a statistically significant decrease in plasma KC/GRO levels compared to vehicle control (Group 2) on Days 5, 7 and 21 (p<0.05, by student's t test) as indicated in Table 6. Overall, the decrease of a plasma biomarker of inflammation corroborates the ability of compound 175 to ameliorate tendonitis.











TABLE 6









Plasma KC/GRO Concentration (pg/mL)










Vehicle
Compound 175-Treated Groups













Group 2
Group 3
Group 4
Group 5
Group 6



0
3 mg/mL
10 mg/mL
3 mg/mL
10 mg/mL










Day
0.5% Phx
0.5% BA
0.5% Phx















0
219 ± 94.9
78.2 ± 14.9 
224 ± 40.1
112 ± 74.0
254 ± 60.6


1
450 ± 53.1
472 ± 47.2
360 ± 31.4
416 ± 47.9
375 ± 44.4


3
451 ± 50.2
449 ± 53.9
454 ± 52.2
374 ± 89.1
405 ± 42.1


5
778 ± 56.9
561 ± 97.2
 327 ± 49.9**
 431 ± 24.5**
 328 ± 37.1**


7
708 ± 95.5
445 ± 54.5
396 ± 92.4
422 ± 47.9
 299 ± 53.9*


14
704 ± 170 
319 ± 57.7
343 ± 59.4
505 ± 41.0
265 ± 20.8


21
488 ± 98.8
 150 ± 53.8*
350 ± 25.6
226 ± 37.3
342 ± 39.7





*p < 0.05 (student's t test) on Day 7 (Group 6) and on Day 21 (Group 3) compared to Group 2 (Vehicle) on Days 7 and 21.


**p < 0.01 (student's t test) on Day 5 compound 175-treated groups (Groups 4, 5 and 6) compared to Group 2 (Vehicle).






Tendinopathy is an acute injury of the tendon that involves inflammation and tendon damage. If left untreated, repeated injury can lead to tendon ruptures and require surgery. Both doses of compound 175 (3 and 10 mg/ml with either BA or Phx preservatives) were efficacious in the rat tendonitis model. Based on the foregoing results, neither preservative, BA or Phx, appeared to improve the efficacy of compound 175. The results demonstrate that treatment with compound 175 ameliorated tendinopathy as assessed by blinded histological scoring of the tendon. To further confirm this finding, it has been shown that compound 175 also results in a decrease of an inflammatory plasma biomarker, KC/GRO.


Example 14

Pharmacokinetics


Thirty (30) naïve male Sprague Dawley rats were divided into three dose groups (Groups 1-3; 10 animals/group). compound 175 was formulated in 1% hydroxypropyl methylcellulose (HPMC) 40-0101/20% propylene glycol at 1 or 10 mg/mL containing 1% benzyl alcohol (BA) as preservative (Groups 1 and 2) or at 10 mg/mL containing 0.5% Tween 80 (used in earlier rat studies) without BA preservative (Group 3). The compound 175 formulation was applied once topically to each ankle of the hind leg of each animal at 60 μL volume (30 μL/cm2×2 cm2) over the area of the Achilles tendon. The study design is presented in Table 7 below.













TABLE 7









Cmpd

Left and Right Hind limba
















175

Dose

Dose per
Total Dose




Conc.
Preser-
Volume
Dose Area
Unit Area
Volume


Group
N/Sex
(mg/mL)
vativeb
(μl/cm2)
(cm2)
(mg/cm2)
(μL)

















1
10/M
1
BA
30
2
0.03
60


2
10/M
10



0.3


3
10/M
10
No


0.3





preser-





vative






aTopical application to left and right ankles of the hind limbs over the area of the Achilles tendon




bCompound 175 topical formulations: 1% HPMC 40-0101/20% propylene glycol containing 1% benzyl alcohol (BA) as preservative (Groups 1 and 2), and without BA preservative but with 0.5% Tween 80 (Group 3).







Blood Sample Collection.


Blood samples (˜0.5 mL) were collected via cardiac puncture, into tubes containing K2EDTA as anticoagulant, at 1, 2, 4, 7 and 24 hours post-dose from 2 animals per timepoint (n=2). Plasma was harvested and frozen at −80° C. for bioanalysis.


Tissue Sample Collection.


The Achilles tendons from both ankles were collected following euthanasia at 1, 2, 4, 7 and 24 hours post-dose from 2 animals per timepoint (n=4) from all groups. Skin tissue samples were collected from the dose-site at 7 and 24 hours post-dose from 2 animals per timepoint (n=4) in Group 2. Tissues were frozen at −80° C. for bioanalysis. Blood and tissue collection schedule is outlined in Table 8 below.














TABLE 8






Cmpd.

Blood
Tendon
Skin



175

Collection
Collection
Collection



Conc.
Preser-
Timepoints
Timepoints
Timepoints


Group
(mg/mL)
vative
(h)1)
(h)b
(h)b




















1
1
BA
1, 2, 4,
1, 2, 4,
NS


2
10

7 and 24
7 and 24
7 and 24


3
10
No


NS




preser-




vative





NS = No samples collected.



1)n = 2/timepoint/group for blood collection.




bn = 4/timepoint/group for tendon and skin collection (2 animals per timepoint × 2 ankles).







Experimental Procedures.


Skin tissue samples were processed using the following procedures: 1) Weigh tissue and homogenize using cryoPrep (Covaris); 2) Add 9× volume of Acetonitrile:Methanol (70:30) and vortex for one hour; 3) Aliquot 100 μL of tissue homogenate into 96 microtube polypropylene plate; 4) Add 10 μL of working standard to each standard; 5) Add 10 μL of methanol in all samples and blanks; 6) Add 10 μL of internal standard (IS) to all tubes except blanks; 7) Spin in centrifuge at 3000 rpm for 10 minutes; and 8) Transfer 100 μL of supernatant into 96-well plate containing 150 μL of water. Cap and vortex for LC/MS/MS analysis.


Bioanalytical and Pharmacokinetic Analysis.


The chromatograms of the samples were integrated and calibrated using Analyst 1.6.2 (AB Sciex, Redwood City, Calif.). Linear regression with 1/x2 weighting and internal standardization was used for standard calibration, with an acceptance criterion of ±30% of nominal standard concentration. The lower limit of quantitation (LLOQ) was 2.00 ng/mL (plasma), 6.00 ng/g (tendon) and 25.0 ng/g (skin). Pharmacokinetic parameters were estimated by noncompartmental analysis from individual concentration versus time profiles using Pheonix WinNonlin version 6.3 (Certara, Inc., Princeton, N.J.). The time to maximum concentration (tmax) and the maximum concentration (Cmax) were determined based on measured plasma and tissue concentrations. The area under the curve (AUC0-last) was calculated using the linear trapezoidal rule.


Systemic Exposure.


Systemic exposure to compound 175 was low compared to tendon and skin tissues after a single topical administration of a 1 or 10 mg/mL formulation, with either BA or Tween 80. Plasma exposure in Groups 1 and 2 (1 and 10 mg/mL formulation containing BA) were: 1.30 and 10.6 ng/mL Cmax and 0.650 and 36.1 h·ng/mL AUC(0-last), respectively, showing a dose-proportional increase in Cmax and greater than dose-proportional increase in total exposure (AUC(0-last)). Exposure in Group 3 (10 mg/mL formulation without BA but containing Tween 80) was 1.94 ng/mL Cmax and 6.79 h·ng/mL AUC(0-last), which was ˜0.2× of Group 2 (10 mg/mL with BA).


Tendon Exposure.


Compound 175 Achilles tendon exposure in Groups 1 and 2 (1 and 10 mg/mL formulation containing BA) was 519 and 1930 ng/g Cmax and 2164 and 7270 h·ng/g AUC(0-last), respectively, indicating a less than dose-proportional increase in exposure: 3.4× increase in AUC(0-last) for a 10× dose-increment. Group 3 (10 mg/mL without BA) tendon exposure was 7053 ng/g C. and 28077 h·ng/g AUC(0-last), which was ˜4× of Group 2 (10 mg/mL with BA). Tendon to plasma ratios (AUC(0-last)) were 3329 and 202 in Groups 1 and 2 (1 and 10 mg/mL formulations containing BA) and 4135 in Group 3 (10 mg/mL without BA).


Skin concentrations in Group 2 (10 mg/mL containing BA) were reported as estimated as all values were above quantitation limit (AQL>25000 ng/g). Mean skin concentrations at 7 and 24 hours post-dose were 208600 and 113375 ng/g, roughly 1000× tendon concentrations at these timepoints. Plasma, Tendon and Skin PK parameters are presented below in Table 9 and concentration-time profiles are displayed in FIG. 11.

















TABLE 9













Tissue/



Dose



Cmax

AUC(0-last)
Plasma



Conc.
Preser-


(ng/g

(h · ng/g or
AUC


Group
(mg/mL)
vative
Tissue
tmax (h)
or ng/mL)

last (h)

h · ng/mL)
Ratio























1
1
1% BA
Plasma
1.00
    1.30
1.00
0.650
3329





Tendon
1.00
 519
24.0
2164


2
10
1% BA
Plasma
4.00
   10.6
7.00
36.1
 202





Tendon
1.00
1930
24.0
7270





Skin
7.00
2086001)
24.0
NC
NC


3
10
No
Plasma
7.00
    1.94
7.00
6.79
4135




preserv-
Tendon
1.00
7053
24.0
28077




ative





Total dose volume = 60 μL/ankle of hind limb; n = 2 (plasma), n = 4 (tendon, skin; 2).


NC = not calculated due to lack of sufficient timepoints for AUC calculation.



1)Above quantitation limit (AQL > 25000 ng/g) - value reported as estimated.







Systemic exposure to compound 175 in rats after a single topical administration was low in all dose groups compared to tendon exposure, with tendon to plasma ratios ranging from 202 to 4135 across groups. A dose dependent increase in plasma and tendon exposure was seen between the 1 and 10 mg/mL formulations containing BA. While the 10 mg/mL formulation without BA (containing Tween 80) showed lower systemic exposures (0.2×) compared to the formulation containing BA, the tendon exposure was 4 fold higher than the 10 mg/mL formulation containing BA.


The term “comprising” as used herein is synonymous with “including,” “containing,” or “characterized by,” and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.

Claims
  • 1. A method of treating tendinopathy, the method comprising administering to a subject (e.g., a subject in need thereof) a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof having the structure of:
  • 2. The method of claim 1, wherein the tendinopathy is tendinosis.
  • 3. The method of claim 1, wherein the tendinopathy is tendinitis.
  • 4. The method of claim 1, wherein the tendinopathy is tenosynovitis.
RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 62/411,478, filed Oct. 21, 2016, which is incorporated herein by reference in its entirety.

US Referenced Citations (186)
Number Name Date Kind
4164559 Miyata et al. Aug 1979 A
4474752 Haslam et al. Oct 1984 A
4603139 King Jul 1986 A
5037844 Hamminga et al. Aug 1991 A
5922733 Forbes et al. Jul 1999 A
6120484 Silverstein Sep 2000 A
6358978 Ritzeler et al. Mar 2002 B1
6377849 Lenarz et al. Apr 2002 B1
6440102 Arenberg et al. Aug 2002 B1
6555539 Reich et al. Apr 2003 B2
6648873 Arenberg et al. Nov 2003 B2
6831175 Li et al. Dec 2004 B2
6884890 Kania et al. Apr 2005 B2
6897208 Edwards et al. May 2005 B2
6911211 Eini et al. Jun 2005 B2
6919461 Reich et al. Jul 2005 B2
7008953 Kephart et al. Mar 2006 B2
7064215 Renhowe et al. Jun 2006 B2
7232912 Reich et al. Jun 2007 B2
7285565 Zhu et al. Oct 2007 B2
7390815 Davies et al. Jun 2008 B2
7429609 Ohi et al. Sep 2008 B2
7452993 Arnold et al. Nov 2008 B2
7468376 Rosales et al. Dec 2008 B2
7482342 D'Orchymont et al. Jan 2009 B2
7488737 Xie et al. Feb 2009 B2
7491710 Cherrier et al. Feb 2009 B2
7541367 Chin et al. Jun 2009 B2
7626021 Arnold et al. Dec 2009 B2
7642278 Jansen et al. Jan 2010 B2
7666867 Makriyannis et al. Feb 2010 B2
7812043 Lau et al. Oct 2010 B2
7829558 Arnold et al. Nov 2010 B2
7842711 D'Orchymont et al. Nov 2010 B2
7902217 Xie et al. Mar 2011 B2
7943616 Cox et al. May 2011 B2
8008481 Ericsson et al. Aug 2011 B2
8088772 Garcia et al. Jan 2012 B2
8129519 Cholody et al. Mar 2012 B2
8158647 Blaney et al. Apr 2012 B2
8252812 Hood et al. Aug 2012 B2
8288425 Edwards et al. Oct 2012 B2
8304408 Wrasidlo et al. Nov 2012 B2
8450340 Hood et al. May 2013 B2
8604052 Hood et al. Dec 2013 B2
8618128 Hood et al. Dec 2013 B1
8637508 Badiger et al. Jan 2014 B2
8664241 Hood et al. Mar 2014 B2
8673936 Hood et al. Mar 2014 B2
8697887 Hood et al. Apr 2014 B2
8703794 Hood et al. Apr 2014 B2
8815897 Hood et al. Aug 2014 B2
8822478 Hood et al. Sep 2014 B2
8846714 Hood et al. Sep 2014 B2
8883822 Hood et al. Nov 2014 B2
8901150 Hood et al. Dec 2014 B2
8987298 Hood et al. Mar 2015 B2
9012472 Hood et al. Apr 2015 B2
9056874 Adams et al. Jun 2015 B2
9067939 Hood et al. Jun 2015 B2
9090613 Hood et al. Jul 2015 B2
9174967 Korber et al. Nov 2015 B2
9199991 Hood et al. Dec 2015 B2
9221793 Hood et al. Dec 2015 B2
9233104 Hood et al. Jan 2016 B2
9381192 Hood et al. Jul 2016 B2
9538272 Auclair et al. Jan 2017 B2
9540398 KC et al. Jan 2017 B2
9586977 Hood et al. Mar 2017 B2
9745271 Hood et al. Aug 2017 B2
9763927 Hood et al. Sep 2017 B2
9763951 Kumar et al. Sep 2017 B2
9802916 Hood et al. Oct 2017 B2
9815854 Kumar et al. Nov 2017 B2
9828372 Kumar et al. Nov 2017 B2
9844536 Kumar et al. Dec 2017 B2
9855272 Hood et al. Jan 2018 B2
9889140 Kc et al. Feb 2018 B2
10407425 Hood et al. Sep 2019 B2
20020103229 Bhagwat et al. Aug 2002 A1
20020161022 Reich et al. Oct 2002 A1
20030199516 Moser et al. Oct 2003 A1
20040048868 Edwards et al. Mar 2004 A1
20040077681 Rawlings et al. Apr 2004 A1
20040176325 Munson et al. Sep 2004 A1
20040236101 Makriyannis et al. Nov 2004 A1
20050009894 Babin et al. Jan 2005 A1
20050026960 Kephart et al. Feb 2005 A1
20050070546 Arrington et al. Mar 2005 A1
20050090529 McAlpine et al. Apr 2005 A1
20050192262 Hagstrom et al. Sep 2005 A1
20050208582 Ohi et al. Sep 2005 A1
20050261339 Ohi et al. Nov 2005 A1
20060004000 D'Orchymont et al. Jan 2006 A1
20060014756 Edwards et al. Jan 2006 A1
20060079564 Jansen et al. Apr 2006 A1
20060094706 Paruch et al. May 2006 A1
20060111322 Reich et al. May 2006 A1
20060116519 Ma et al. Jun 2006 A1
20060135589 Berdino et al. Jun 2006 A1
20060142345 Kephart et al. Jun 2006 A1
20060167056 Rynberg et al. Jul 2006 A1
20060264897 Lobl Nov 2006 A1
20070027140 Lau et al. Feb 2007 A1
20070049598 Billedeau et al. Mar 2007 A1
20070060616 Bennett et al. Mar 2007 A1
20070078147 Schumacher et al. Apr 2007 A1
20070185187 D'Orchymont et al. Aug 2007 A1
20070219257 Beachy et al. Sep 2007 A1
20070282101 Ericsson et al. Dec 2007 A1
20080004270 Gill et al. Jan 2008 A1
20080132495 Berdini et al. Jun 2008 A1
20080255085 Arvidsson et al. Oct 2008 A1
20080262205 Haar et al. Oct 2008 A1
20080287452 Bursavich et al. Nov 2008 A1
20090005356 Blaney et al. Jan 2009 A1
20090005377 Almansa Rosales et al. Jan 2009 A1
20090048249 Chiu et al. Feb 2009 A1
20090054397 Ohi et al. Feb 2009 A1
20090099062 Lee et al. Apr 2009 A1
20090203690 Akritopoulou-Zanze et al. Aug 2009 A1
20090247504 Churcher et al. Oct 2009 A1
20090264446 Rosales et al. Oct 2009 A9
20090286983 Almansa Rosales et al. Nov 2009 A1
20100280063 Price et al. Nov 2010 A1
20100298377 Aletru et al. Nov 2010 A1
20110009353 Chen-Kiang et al. Jan 2011 A1
20110021467 D'Orchymont et al. Jan 2011 A1
20110034441 Hood et al. Feb 2011 A1
20110034497 Hood et al. Feb 2011 A1
20110082144 Lau et al. Apr 2011 A1
20110178075 Xie et al. Jul 2011 A1
20110190290 Hood et al. Aug 2011 A1
20120053345 Ericson et al. Mar 2012 A1
20120059047 Prins et al. Mar 2012 A1
20120129837 Cholody et al. May 2012 A1
20120277229 Bearss et al. Nov 2012 A1
20130039906 Do et al. Feb 2013 A1
20130079329 Hood Mar 2013 A1
20130267548 Follmann et al. Oct 2013 A1
20140194441 K.C. et al. Jul 2014 A1
20140263319 Fazi et al. Sep 2014 A1
20150087687 Brown Mar 2015 A1
20160068529 K.C. et al. Mar 2016 A1
20160068547 K.C. et al. Mar 2016 A1
20160068548 K.C. et al. Mar 2016 A1
20160068549 K.C. et al. Mar 2016 A1
20160068550 K.C. et al. Mar 2016 A1
20160068551 K.C. et al. Mar 2016 A1
20160075701 K.C. et al. Mar 2016 A1
20160090380 K.C. et al. Mar 2016 A1
20160101092 Hood et al. Apr 2016 A1
20160297812 Hood et al. Oct 2016 A1
20170333409 Hood et al. Nov 2017 A1
20170349584 Kumar et al. Dec 2017 A1
20180086754 Kumar et al. Mar 2018 A1
20180133199 Dellamary May 2018 A1
20180141963 Kumar et al. May 2018 A1
20180148444 Kumar et al. May 2018 A1
20180153873 Hood et al. Jun 2018 A1
20180162840 Kumar et al. Jun 2018 A1
20180177787 Kumar et al. Jun 2018 A1
20180201624 Kumar et al. Jul 2018 A1
20180207141 Kumar et al. Jul 2018 A1
20180214427 KC et al. Aug 2018 A1
20180214428 KC et al. Aug 2018 A1
20180214429 KC et al. Aug 2018 A1
20180215753 KC et al. Aug 2018 A1
20180221341 KC et al. Aug 2018 A1
20180221350 KC et al. Aug 2018 A1
20180221351 KC et al. Aug 2018 A1
20180221352 KC et al. Aug 2018 A1
20180221353 KC et al. Aug 2018 A1
20180221354 KC et al. Aug 2018 A1
20180222891 KC et al. Aug 2018 A1
20180222923 KC et al. Aug 2018 A1
20180228780 KC et al. Aug 2018 A1
20180228781 KC et al. Aug 2018 A1
20180228782 KC et al. Aug 2018 A1
20180228783 KC et al. Aug 2018 A1
20180228784 KC et al. Aug 2018 A1
20180228785 KC et al. Aug 2018 A1
20180230142 KC et al. Aug 2018 A1
20180237416 Hood et al. Aug 2018 A1
20180250269 KC et al. Sep 2018 A1
20180256588 Hood et al. Sep 2018 A1
Foreign Referenced Citations (77)
Number Date Country
1394205 Jan 2003 CN
1671710 Sep 2005 CN
1829713 Sep 2006 CN
101440092 May 2009 CN
20122 Jan 2010 KZ
2331640 Aug 2008 RU
WO1987005297 Sep 1987 WO
WO1996002537 Feb 1996 WO
WO2001002369 Jan 2001 WO
WO2001053268 Jul 2001 WO
WO2003004488 Jan 2003 WO
WO2003035005 May 2003 WO
WO2003035065 May 2003 WO
WO2003035644 May 2003 WO
WO2003051366 Jun 2003 WO
WO2003070236 Aug 2003 WO
WO2003070706 Aug 2003 WO
WO2003097610 Nov 2003 WO
WO2003101968 Dec 2003 WO
WO2003101993 Dec 2003 WO
WO2004014864 Feb 2004 WO
WO2004031158 Apr 2004 WO
WO2004076450 Sep 2004 WO
WO2005009997 Feb 2005 WO
WO2005012301 Feb 2005 WO
WO2005014554 Feb 2005 WO
WO2005047266 May 2005 WO
WO2005049019 Jun 2005 WO
WO2005092890 Oct 2005 WO
WO2005099703 Oct 2005 WO
WO2005110410 Nov 2005 WO
WO2006001894 Jan 2006 WO
WO2006015124 Feb 2006 WO
WO2006024945 Mar 2006 WO
WO2006054143 May 2006 WO
WO2006054151 May 2006 WO
WO2006063302 Jun 2006 WO
WO2006063841 Jun 2006 WO
WO2006130673 Dec 2006 WO
WO2007061360 May 2007 WO
WO2007107346 Sep 2007 WO
WO2007117465 Oct 2007 WO
WO2007147874 Dec 2007 WO
WO2008061109 May 2008 WO
WO2008071397 Jun 2008 WO
WO2008071398 Jun 2008 WO
WO2008071451 Jun 2008 WO
WO2008124848 Oct 2008 WO
WO2008137408 Nov 2008 WO
WO2008140792 Nov 2008 WO
WO2008147713 Dec 2008 WO
WO2008150914 Dec 2008 WO
WO2008154241 Dec 2008 WO
WO2008156757 Dec 2008 WO
WO2009011850 Jan 2009 WO
WO2009016072 Feb 2009 WO
WO2009029609 Mar 2009 WO
WO2009061345 May 2009 WO
WO2010064875 Jun 2010 WO
WO2010107765 Sep 2010 WO
WO2010111060 Sep 2010 WO
WO2010132725 Nov 2010 WO
WO2011011722 Jan 2011 WO
WO2011019648 Feb 2011 WO
WO2011019651 Feb 2011 WO
WO2011050245 Apr 2011 WO
WO2011079076 Jun 2011 WO
WO2011084486 Jul 2011 WO
WO2011123890 Oct 2011 WO
WO2012068589 May 2012 WO
WO2012104388 Aug 2012 WO
WO2012129562 Sep 2012 WO
WO2013024011 Feb 2013 WO
WO 2013040215 Mar 2013 WO
WO2013030138 Mar 2013 WO
WO2013113722 Aug 2013 WO
WO2017079765 May 2017 WO
Non-Patent Literature Citations (279)
Entry
Bass. Tendinopathy: why the difference between tendinitis and tendinosis matters. International Jouranl of Therapeutic Massage and Bodywork. vol. 5, No. 1, Mar. 2012.
U.S. Appl. No. 16/015,996, filed Jun. 22, 2018, Kumar KC et al.
“Application of Hamish Christopher Swan Wood, Norman Whittaker, Irene Stirling and Kyuji Ohta.,” 582 F.2d 638 (Fed. Cir. 1978), 2 pages.
Adaimy et al., “Mutation in WNT10A is Associated with an Autosomal Recessive Ectodermal Dysplasia: The Odonto-onycho-dermal Dysplasia,” Am. J. Hum. Genet., (Oct. 2007), 81(4), 821-828.
Anastas and Moon, “WNT signalling pathways as therapeutic targets in cancer,” Nat Rev Cancer, 13(1):11-26, Jan. 2013.
Andres, “Molecular genetics and animal models in autistic disorder,” Brain Research Bulletin, (2002), 57(1), 109-119.
Barker and Clevers, “Mining the Wnt pathway for cancer therapeutics,” Nat Rev Drug Discov., 5(12):997-1014, Dec. 2006.
Beyer et al., “Extended report: β-catenin is a central mediator of pro-fibrotic Wnt signaling in systemic sclerosis,” Ann Rheum Dis, 71:761-767, online Feb. 2012.
Biason-Lauber et al., “A WNT4 Mutation Associated with Müllerian-Duct Regression and Virilization in a 46,XX Woman,” N. Engl. J. Med., (Aug. 2004), 351(8), 792-798.
Blaydon et al., “The gene encoding R-spondin 4 (RSPO4), a secreted protein implicated in Wnt signaling, is mutated in inherited anonychia,” Nat. Genet., (Nov. 2006), 38(11), 1245-1247.
Blom et al., “Involvement of the Wnt signaling pathway in experimental and human osteoarthritis: prominent role of Wnt-induced signaling protein 1,” Arthritis Rheum., 60(2):501-512, Feb. 2009.
Boyden et al., “High Bone Density Due to a Mutation in LDL-Receptor—Related Protein 5,” N. Engl. J. Med., (May 2002), 346(20):1513-1521.
Brack et al., “Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis,” Science., 317(5839):807-810, Aug. 2007.
Brown et al., “Toxicity and toxicokinetics of the cyclin-dependent kinase inhibitor AG-024322 in cynomolgus monkeys following intravenous infusion,” Cancer Chemother Pharmacol., 62(6):1091-1101, Epub May 2008.
Chilosi et al., “The pathogenesis of COPD and IPF: Distinct horns of the same devil?,” Respiratory Research, 13:3, 2012.
Chockalingam et al., “Elevated aggrecanase activity in a rat model of joint injury is attenuated by an aggrecanase specific inhibitor,” Osteoarthritis Cartilage, Mar. 2011, 19(3): 315-323.
Chou and Talalay, “Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors,” Advances in Enzyme Regulation (1984), 22, 27-55.
Chou, “Drug combination studies and their synergy quantification using the Chou-Talalay method,” Cancer Res., 70(2):440-446, Jan. 2010.
Chou, “Graphic rule for drug metabolism systems,” Current Drug Metabolism, (May 2010) 11(4):369-378.
Christodoulides et al., “WNT10B mutations in human obesity,” Diabetologia, (2006) 49(4):678-684.
Clevers and Nusse, “Wnt/β-catenin signaling and disease,” Cell, (Jun. 2012), 149(6):1192-1205.
Clevers, “Wnt/beta-catenin signaling in development and disease,” Cell, (Nov. 2006), 127(3), 469-480.
Corr, “Wnt-beta-catenin signaling in the pathogenesis of osteoarthritis,” Nat Clin Pract Rheumatol., 4(10):550-556, Oct. 2008.
D'Alessio et al., “Benzodipyrazoles: a new class of potent CDK2 inhibitors,” Bioorganic & Medicinal Chemistry Letters (2005), 15(5), 1315-1319.
Dann et al., “Insights into Wnt binding and signaling from the structures of two Frizzled cysteine-rich domains,” Nature, (Jul. 2001), 412, pp. 86-90.
Datta et al., “Novel therapeutic approaches for pulmonary fibrosis,” Br J Pharmacol., 163(1):141-172, May 2011.
Davidson et al., “Emerging links between CDK cell cycle regulators and Wnt signaling,” Trends Cell Biol., Aug. 2010, 20(8):453-460.
De Ferrari and Inestrosa, “Wnt signaling function in Alzheimer's disease,” Brain Research Reviews,(2000), 33(1): 1-12.
De Ferrari and Moon, “The ups and downs of Wnt signaling in prevalent neurological disorders,” Oncogene, (2006) 25(57): 7545-7553.
De Ferrari et al., “Common genetic variation within the Low-Density Lipoprotein Receptor-Related Protein 6 and late-onset Alzheimer's disease,” Proc. Natl. Acad. Sci. USA, (May 2007), 104(22):9434-9439.
Dermer, “Another Anniversary for the War on Cancer,” Nature Biotechnology, 12:320 (1994).
Dessalew et al., “3D-QSAR CoMFA and CoMSIA study on benzodipyrazoles as cyclin dependent kinase 2 inhibitors,” Medicinal Chemistry, (2008), 4(4), 313-321.
Dong et al., “QSAR study of Akt/protein kinase B (PKB) inhibitors using support vector machine,” European Journal of Medicinal Chemistry, (Oct. 2009), pp. 44(10): 4090-4097.
du Bois, “Strategies for treating idiopathic pulmonary fibrosis,” Nature Reviews Drug Discovery, 9(2): 129-140 (Feb. 2010).
Edamoto et al., “Alterations of RB1, p53 and Wnt pathways in hepatocellular carcinomas associated with hepatitis C, hepatitis B and alcoholic liver cirrhosis,” Int J Cancer., 106(3):334-341, Sep. 1, 2003.
Egloff et al., “Gastrin-releasing peptide receptor expression in non-cancerous bronchial epithelia is associated with lung cancer: a case-control study,” Respiratory Research, 13:9, Feb. 2012.
Espada et al., “Wnt signalling and cancer stem cells,” Clin. Transl. Oncol., (2009), 11(7), 411-27.
Ewan et al., “A useful approach to identify novel small-molecule inhibitors of Wnt-dependent transcription,” Cancer Res. (2010), 70(14), 5963-5973.
Florez et al., “TCF7L2 Polymorphisms and Progression to Diabetes in the Diabetes Prevention Program,” N. Engl. J. Med., (Jul. 2006), 355(3):241-250.
Freese et al., “Wnt signaling in development and disease,” Neurobiology of Disease, (2010) 38(2):148-153.
Fujii et al., “An antagonist of dishevelled protein-protein interaction suppresses beta-catenin-dependent tumor cell growth,” Cancer Res., 67(2):573-579, Jan. 2007.
Fukuzawa et al., “Beckwith-Wiedemann Syndrome-associated Hepatoblastoma: Wnt Signal Activation Occurs Later in Tumorigenesis in Patients with 11p15.5 Uniparental Disomy,” Pediatric and Developmental Pathology (2003), 6(4): 299-306.
Giles et al., “Caught up in a Wnt storm: Wnt signaling in cancer,” Biochim Biophys Acta., 1653(1):1-24, Jun. 2003.
Handeli and Simon, “A small-molecule inhibitor of Tcf/beta-catenin signaling down-regulates PPARgamma and PPARdelta activities,” Mol Cancer Ther., 7(3):521-529, Mar. 2008.
Henderson Jr. et al., “Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis,” Proc Natl Acad Sci U S A., 107(32):14309-14314, Epub Jul. 2010.
Hu et al., “Discovery of indazoles as inhibitors of Tp12 kinase,” Bioorganic & Medicinal Chemistry Letters, (Aug. 2011) 21(16): 4758-4761.
Huang et al., “Tankyrase inhibition stabilizes axin and antagonizes Wnt signaling,” Nature, (Oct. 2009), 461(7264): 614-620.
Huang et al., “Synthesis of 3-(1H-benzimidazol-2-yl)-5-isoquinolin-4-ylpyrazolo[1,2-b]pyridine, a potent cyclin dependent kinase 1 (CDK1) inhibitor,” Bioorganic & Medicinal Chemistry Letters, (2007) 17(5): 1243-1245.
Hübner et al., “Standardized quantification of pulmonary fibrosis in histological samples,” Biotechniques, 44(4):507-511, 514-517, Apr. 2008.
Im et al., “Wnt inhibitors enhance chondrogenesis of human mesenchymal stem cells in a long-term pellet culture,” Biotechnol Lett., 33(5):1061-1068, Epub Jan. 2011.
Inestrosa and Toledo, “The role of Wnt signaling in neuronal dysfunction in Alzheimer's Disease,” Mol Neurodegener, 3:9, doi:10.1186/1750-1326-3-9, 13 pages, Jul. 2008.
International Search Report and Written Opinion for International Application No. PCT/US2017/057536, dated Feb. 8, 2018, 15 pages.
Invitation to Pay Additional Fees for Application No. PCT/US2017/057536, dated Dec. 14, 2017, 11 pages.
Janssens et al., “The Wnt-dependent signaling pathways as target in oncology drug discovery,” Invest New Drugs., 24(4):263-280, Jul. 2006.
Jenkins et al., “Germline mutations in WTX cause a sclerosing skeletal dysplasia but do not predispose to tumorigenesis,” Nat. Genet. (Jan. 2009), 41(1), 95-100.
Jessen et al., “Peripheral white blood cell toxicity induced by broad spectrum cyclin-dependent kinase inhibitors,” Journal of Applied Toxicology (Jan. 2007), 27(2), 133-142.
Johnson et al., “A stem cell-based approach to cartilage repair,” Science., 336(6082):717-721, Epub Apr. 5, 2012.
Kanazawa et al., “Association of the Gene Encoding Wingless-Type Mammary Tumor Virus Integration-Site Family Member 5B (Wntsb) with Type 2 Diabetes,” Am. J. Hum. Genet. (2004), 75(5), 832-843.
Karlberg et al., “Structural basis for the interaction between tankyrase-2 and a potent Wnt-signaling inhibitor,” J. Med. Chem. (2010), 53(14), 5352-5.
Kibar et al., “Mutations in VANGL1 Associated with Neural-Tube Defects,” N. Engl. J. Med., (Apr. 2007), 356(14):1432-1437.
King et al., “BUILD-3: a randomized, controlled trial of bosentan in idiopathic pulmonary fibrosis,” Am J Respir Crit Care Med., 184(1):92-99, Epub Apr. 2011.
Kishimoto et al: “Wnt/Beta-Catenin Signaling Suppresses Expressions of Ses, Mkx and Tnmd in Tendon-Derived Cells,” Plos One, Jul. 27, 2017, 12(7), E0182051, pp. 1-17.
Kuwajima et al., “Necdin Promotes GABAergic Neuron Differentiation in Cooperation with Dlx Homeodomain Proteins,” Journal of Neuroscience (May 2006), 26(20), 5383-5392.
Lacy et al., “Generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig(TM)) molecule that specifically and potently neutralizes both IL-1α and IL-1β,” Mabs, May 2015, 7(3): 605-619.
Lammi et al., “Mutations in AXIN2 Cause Familial Tooth Agenesis and Predispose to Colorectal Cancer,” Am. J. Hum. Genet. (2004), 74(5), 1043-1050.
Leyns et al., “Frzb-1 is a Secreted Antagonist of Wnt Signaling Expressed in the Spemann Organizer,” Cell (Mar. 1997), 88(6), 747-756.
Li et al., “Artesunate attenuates the growth of human colorectal carcinoma and inhibits hyperactive Wnt/beta-catenin pathway,” Int J Cancer., 121(6):1360-1365, Sep. 2007.
Lin et al., “Synthesis and evaluation of pyrazolo[3,4-b]pyridine CDK1 inhibitors as anti-tumor agents,” Bioorganic & Medicinal Chemistry Letters, (Aug. 2007), 17(15): 4297-4302.
Liu, et.al., “Fibrotic lung fibroblasts show blunted inhibition by cAMP due to deficient cAMP response element-binding protein phosphorylation,” J Pharmacol Exp Ther., 315(2):678-687, Epub Aug. 3, 2005.
Lories et al., “To Wnt or not to Wnt: the bone and joint health dilemma,” Nat Rev Rheumatol., 9(6):328-339, Epub Mar. 2013.
Low et al., “Phenotypic fingerprinting of small molecule cell cycle kinase inhibitors for drug discovery,” Curr Chem Genomics., 3:13-21, Mar. 2009.
Lu et al., “Structure-activity relationship studies of small-molecule inhibitors of Wnt response,” Bioorganic & Medicinal Chemistry Letters, (Jul. 2009), 19(14):3825-3827.
Lui: “Histopathological Changes in Tendinopathypotential Roles of BMPs?” Rheumatology, May 2013, 52:2116-2126.
Luo et al., “Fragile X Mental Retardation Protein Regulates Proliferation and Differentiation of Adult Neural Stem/Progenitor Cells,” PLoS Genetics, (Apr. 2010), 6(4):e1000898, 15 pages.
Luu et al., “Wnt/beta-catenin signaling pathway as a novel cancer drug target,” Curr Cancer Drug Targets., 4(8):653-671, Dec. 2004.
Luyten et al., “Wnt signaling and osteoarthritis,” Bone, 44(4):522-527, Epub Dec. 14, 2008.
MacDonald et al., “Wnt/beta-catenin signaling: components, mechanisms, and diseases,” Dev. Cell (Jul. 2009), 17(1), 9-26.
Mandel et al., “SERKAL Syndrome: an Autosomal-Recessive Disorder Caused by a Loss-of-Function Mutation in WNT4,” Am. J. Hum. Genet., (Jan. 2008), 82(1), 39-47.
Mani, et al., “LRP6 Mutation in a Family with Early Coronary Disease and Metabolic Risk Factors,” Science, (Mar. 2007), 315(5816), 1278-1282.
McBride, et al. “Design and structure-activity relationship of 3-benzimidazol-2-yl-1H-indazoles as inhibitors of receptor tyrosine kinases,” Bioorganic & Medicinal Chemistry Letters (2006), 16(13), 3595-3599.
Misra et al., “1H-Pyrazolo[3,4-b]pyridine inhibitors of cyclin-dependent kinases highly potent 2,6-Difluorophenacyl analogues,” Bioorganic & Medicinal Chemistry Letters, (2003), 13:2405-2408.
Morrisey, “Wnt signaling and pulmonary fibrosis,” Am J Pathol., 162(5):1393-1397, May 2003.
Muddassar et al., “Elucidation of binding mode and three dimensional quantitative structure-activity relationship studies of a novel series of protein kinase B/Akt inhibitors,” Journal of Molecular Modeling, (2009), 15(2): 183-192.
Niemann et al., “Homozygous WNT3 Mutation Causes Tetra-Amelia in a Large Consanguineous Family,” Am. J. Hum. Genet. (2004), 74(3), 558-563.
Nishisho et al., “Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients,” Science, (Aug. 1991), 253(5020):665-669.
Nusse, “Wnt signaling in disease and in development,” Cell Res., 15(1):28-32, Jan. 2005.
Oates et al., “Increased DNA Methylation at the AXIN1 Gene in a Monozygotic Twin from a Pair Discordant for a Caudal Duplication Anomaly,” Am. J. Hum. Genet. (2006 ), 79(1), 155-162.
Oduor et al., “Trypanosoma bracer glycogen synthase kinase-3, a target for anti-trypanosomal drug development: a public-private partnership to identify novel leads,” PLoS Negl Trop Dis., 5(4):e1017, Apr. 2011.
Okerlund and Cheyette, “Synaptic Wnt signaling-a contributor to major psychiatric disorders?” J Neurodev Disord., (2011) 3(2):162-174.
Parsons et al., “Benzo[d]imidazole Transient Receptor Potential Vanilloid 1 Antagonists for the Treatment of Pain: Discovery of trans-2-(2-{2-[2-(4-Trifluoromethyl-phenyl)-vinyl]-1H-benzimidazol-5-yl}-phenyl)-propan-2-ol (Mavatrep),” J Med Chem, May 2015, 58(9): 3859-3874.
Piersanti et al., “Synthesis of benzo[1,2-d;3,4-d′]diimidazole and 1 H-pymzolo[4,3-b]pyridine as putative A2A receptor antagonists,” Organic and Biomolecular Chemistry, Aug. 2007, 5(16):2567-2571.
Polakis, “Wnt signaling and cancer,” Genes Dev., 14: 1837-1851, 2000.
Pubchem. Substance Record for SID 164345938. Deposit Date: Nov. 4, 2013. [retrieved on Nov. 16, 2015]. Retrieved from the Internet. <URL: https://pubchem.ncbi.nlm.nih.gov/substance/164345938#section=Top>, 5 pages.
Qin et al. “Complexity of the genotype-phenotype correlation in familial exudative vitreoretinopathy with mutations in the LRP5 and/or FZD4 genes,” Hum. Mutat. (2005), 26(2), 104-112.
Reya and Clevers, “Wnt signalling in stem cells and cancer,” Nature 434: 843-850, Apr. 2005.
Richards et al., “Peripheral blood proteins predict mortality in idiopathic pulmonary fibrosis,” Am J Respir Crit Care Med., 185(1):67-76, Jan. 2012.
Rivera et al., “An X Chromosome Gene, WTX, is Commonly Inactivated in Wilms Tumor,” Science, (Feb. 2007), 315(5812):642-645, published online Jan. 4, 2007.
Robitaille et al., “Mutant frizzled-4 disrupts retinal angiogenesis in familial exudative vitreoretinopathy,” Nat. Genet., (Sep. 2002), 32(2):326-330.
Rother et al., “Efficacy and safety of epicutaneous ketoprofen in Transfersome (IDEA-033) versus oral celecoxib and placebo in osteoarthritis of the knee: multicentre randomised controlled trial,” Annals of the Rheumatic Diseases, Sep. 2007, 66(9): 1178-1183.
Ryu et al., “Natural derivatives of curcumin attenuate the Wnt/beta-catenin pathway through down-regulation of the transcriptional coactivator p300,” Biochem Biophys Res Commun., 377(4):1304-1308, print Dec. 2008, Epub Nov. 2008.
Salinas, “Wnt signaling in the vertebrate central nervous system: from axon guidance to synaptic function,” Cold Spring Harb Perspect Biol., (2012) 4(2). pii: a008003, 15 pages.
Sato, “Upregulation of the Wnt/beta-catenin pathway induced by transforming growth factor-beta in hypertrophic scars and keloids,” Acta Derm Venereol., 86(4):300-307, 2006.
Seah et al., “Neuronal Death Resulting from Targeted Disruption of the Snf2 Protein ATRX is Mediated by p53,” Journal of Neuroscience (Nov. 2008), 28(47), 12570-12580.
Shih et al., “Pharmacophore modeling and virtual screening to identify potential RET kinase inhibitors,” Bioorg Med Chem Lett., 21(15):4490-4497, Epub Jun. 2011.
Shruster et al., “Wnt signaling enhances neurogenesis and improves neurological function after focal ischemic injury,” PLoS One, (Jul. 2012), 7(7):e40843, 11 pages.
Silva et al, “Advances in Prodrug Design,” Mini-Revs. in Med. Chem. (2005), 5: 893-914.
Solowiej et al., “Characterizing the Effects of the Juxtamembrane Domain on Vascular Endothelial Growth Factor Receptor-2 Enzymatic Activity, Autophosphmylation, and Inhibition by Axitinib,” Biochemistry, (2009), 48(29), 7019-7031.
Staines et al., “Cartilage development and degeneration: a Wnt situation,” Cell Biochem Funct., 30(8):633-642, Epub Jun. 2012.
Sutherland et al., “A robust high-content imaging approach for probing the mechanism of action and phenotypic outcomes of cell-cycle modulators,” Molecular Cancer Therapeutics, (Feb. 2011), 10(2):242-254.
Swaney et al., “A novel, orally active LPA(1) receptor antagonist inhibits lung fibrosis in the mouse bleomycin model,” Br J Pharmacol., 160(7):1699-1713, Aug. 2010.
Takahashi-Yanaga et al., “Celecoxib-induced degradation of T-cell factors-1 and -4 in human colon cancer cells,” Biochem Biophys Res Commun., 377(4):1185-1190, print Dec. 2008, Epub Nov. 2008.
Tamamura et al., “Developmental regulation of Wnt/beta-catenin signals is required for growth plate assembly, cartilage integrity, and endochondral ossification,” J Biol Chem., 280(19):19185-95. Epub Mar. 2005.
Thompson et al., “WNT/beta-catenin signaling in liver health and disease,” Hepatology., 45(5):1298-1305, May 2007.
Trujillo et al., “2-(6-Phenyl-1H-indazol-3-yl)-1H-benzo[d]imidazoles: design and synthesis of a potent and isoform selective PKC-zeta inhibitor,” Bioorg Med Chem Lett., 19(3):908-911, Epub Dec. 6, 2008.
Ugur et al., “Homozygous WNT10b mutation and complex inheritance in Split-Hand/Foot Malformation,” Hum. Mol. Genet. (2008), 17(17), 2644-2653.
Vulpetti et al., “Structure-Based Approaches to Improve Selectivity: CDK2-GSK3β Binding Site Analysis,” Journal of Chemical Information and Modeling (2005), 45(5), 1282-1290.
Wagner et al., “The therapeutic potential of the Wnt signaling pathway in bone disorders,” Curr Mol Pharmacol., 4(1):14-25, Jan. 2011.
Walters and Kleeberger, “Mouse models of bleomycin-induced pulmonary fibrosis,” Current Protocols in Pharmacology, (2008) Chapter 5: Unit 5.46, 1-17.
Wang, et al., “Mutations in X-linked PORCN, a putative regulator of Wnt signaling, cause focal dermal hypoplasia,” Nat. Genet. (Jul. 2007), 39(7), 836-838.
Wantanabe and Dai, “Winning WNT: race to Wnt signaling inhibitors,” Proc Natl Acad Sci U S A.108(15):5929-5930, Epub Mar. 2011.
Watts et.al., “RhoA signaling modulates cyclin D1 expression in human lung fibroblasts; implications for idiopathic pulmonary fibrosis,” Respir Res., 7:88, Jun. 15, 2006.
Weng et al., “Control of Dkk-1 ameliorates chondrocyte apoptosis, cartilage destruction, and subchondral bone deterioration in osteoarthritic knees,” Arthritis Rheum., 62(5):1393-1402, May 2010.
Witherington et al., “5-Aryl-pyrazolo[3,4-b]pyridazines: potent inhibitors of glycogen synthase kinase-3 (GSK-3),” Bioorganic & Medicinal Chemistry Letters, (May 2003), 13(9):1581-1584.
Woods, S. et al., “Mutations in WNT7A Cause a Range of Limb Malformations, Including Fuhrmann Syndrome and Al-Awadi/Raas-Rothschild/Schinzel Phocomelia Syndrome,” Am. J. Hum. Genet. (Aug. 2006), 79(2), 402-408.
Yardy and Brewster, “Wnt signalling and prostate cancer,” Prostate Cancer Prostatic Dis, 8(2):119-126, 2005.
Zhang et al., “Small-molecule synergist of the Wnt/beta-catenin signaling pathway,” Proc Natl Acad Sci U S A., 104(18):7444-7448, Epub Apr. 2007 and correction 104(30):12581, Jul. 2007.
Zhong et al., “Characterization of in vitro and in vivo metabolism of AG-024322, a novel cyclin-dependent kinase (CDK) inhibitor,” Health (2009), 1(4): 249-262.
Zhu et al. “Design and synthesis of pyridine-pyrazolopyridine-based inhibitors of protein kinase B/Akt,” Bioorganic & Medicinal Chemistry, Mar. 2007, 15(6):2441-2452.
U.S. Appl. No. 12/852,681, filed Aug. 9, 2010, Hood et al.
U.S. Appl. No. 12/968,505, filed Dec. 15, 2010, Hood et al.
U.S. Appl. No. 13/855,874, filed Apr. 3, 2013, Hood et al.
U.S. Appl. No. 13/938,692, filed Jul. 10, 2013, Hood et al.
U.S. Appl. No. 14/331,427, filed Jul. 15, 2014, Hood et al.
U.S. Appl. No. 14/465,056, filed Aug. 21, 2014, Hood et al.
U.S. Appl. No. 14/718,354, filed May 21, 2015, Hood et al.
U.S. Appl. No. 15/244,687, filed Aug. 23, 2016, Hood et al.
U.S. Appl. No. 15/812,629, filed Nov. 14, 2017, Hood et al.
U.S. Appl. No. 12/852,706, filed Aug. 9, 2010, Hood et al.
U.S. Appl. No. 13/552,188, filed Jul. 18, 2012, Hood et al.
U.S. Appl. No. 13/938,691, filed Jul. 10, 2013, Hood et al.
U.S. Appl. No. 14/019,103, filed Sep. 5, 2013, Hood et al.
U.S. Appl. No. 14/334,005, filed Jul. 17, 2014, Hood et al.
U.S. Appl. No. 14/741,645, filed Jun. 17, 2015, Hood et al.
U.S. Appl. No. 15/184,553, filed Jun. 16, 2016, Hood et al.
U.S. Appl. No. 15/681,035, filed Aug. 18, 2017, Hood et al.
U.S. Appl. No. 13/614,296, filed Sep. 13, 2012, Hood et al.
U.S. Appl. No. 14/019,229, filed Sep. 5, 2013, Hood et al.
U.S. Appl. No. 14/940,958, filed Nov. 13, 2015, Hood et al.
U.S. Appl. No. 15/709,057, filed Sep. 19, 2017, Hood et al.
U.S. Appl. No. 13/800,963, filed Mar. 13, 2013, Hood et al.
U.S. Appl. No. 14/019,940, filed Sep. 6, 2013, Hood et al.
U.S. Appl. No. 14/178,749, filed Feb. 12, 2014, Hood et al.
U.S. Appl. No. 14/621,195, filed Feb. 12, 2015, Hood et al.
U.S. Appl. No. 14/939,434, filed Nov. 12, 2015, Hood et al.
U.S. Appl. No. 15/968,555, filed May 1, 2018, Hood et al.
U.S. Appl. No. 13/887,177, filed May 3, 2013, Hood et al.
U.S. Appl. No. 14/019,147, filed Sep. 5, 2013, Hood et al.
U.S. Appl. No. 14/454,279, filed Aug. 7, 2014, Hood et al.
U.S. Appl. No. 14/621,222, filed Feb. 12, 2015, Hood et al.
U.S. Appl. No. 14/962,681, filed Dec. 8, 2015, Hood et al.
U.S. Appl. No. 15/420,398, filed Jan. 31, 2017, Hood et al.
U.S. Appl. No. 14/149,948, filed Jan. 8, 2014, Kumar KC et al.
U.S. Appl. No. 15/889,403, filed Feb. 6, 2018, Kumar KC et al.
U.S. Appl. No. 14/847,259, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/298,346, filed Oct. 20, 2016, Kumar KC et al.
U.S. Appl. No. 15/716,803, filed Sep. 27, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,336, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/661,231, filed Jul. 27, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,299, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/591,566, filed May 10, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,287, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/363,086, filed Nov. 29, 2016, Kumar KC et al.
U.S. Appl. No. 15/808,602, filed Nov. 9, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,344, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/257,398, filed Sep. 6, 2016, Kumar KC et al.
U.S. Appl. No. 15/673,834, filed Aug. 10, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,394, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/357,494, filed Nov. 21, 2016, Kumar KC et al.
U.S. Appl. No. 15/716,894, filed Sep. 27, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,371, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/267,939, filed Sep. 16, 2016, Kumar KC et al.
U.S. Appl. No. 15/843,818, filed Dec. 15, 2017, Kumar KC et al.
U.S. Appl. No. 14/847,379, filed Sep. 8, 2015, Kumar KC et al.
U.S. Appl. No. 15/668,992, filed Aug. 4, 2017, Kumar KC et al.
U.S. Appl. No. 15/749,587, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,586, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,592, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,606, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,608, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,701, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,706, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,713, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,718, filed Feb. 1, 2018 Kumar KC et al.
U.S. Appl. No. 15/749,721, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,741, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,727, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,739, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,737, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,742, filed Feb. 1, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,868, filed Feb. 2, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,929, filed Feb. 2, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,910, filed Feb. 2, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,923, filed Feb. 2, 2018, Kumar KC et al.
U.S. Appl. No. 15/749,922, filed Feb. 2, 2018, Kumar KC et al.
U.S. Appl. No. 15/611,150, filed Jun. 1, 2017, Kumar KC.
U.S. Appl. No. 15/806,321, filed Nov. 7, 2017, Dellamary.
Cecil Textbook of Medicine, edited by Bennet, J.C., and Plum F., 20th edition, vol. 1, 1004-1010, 1996.
Dermer et al., “Another Anniversary for the War on Cancer,” Bio/Technology, Mar. 1994, 12:320.
Freshney et al., Culture of Animal Cells, A Manual of Basic Technique, Alan R. Liss, Inc., 1983, New York, p. 1-6.
Golub et al., “Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring,” Science, Oct. 1999, 286(5439):531-537.
Yamada et al., “Emergence of TNIK inhibitors in cancer therapeutics,” Cancer Sci, May 2017, 108(5):818-823.
Adult Brain Tumors Treatment, National Cancer Institute, pp. 1-21 (Jan. 24, 2013), 21 pages.
Ai et al., “Optimal Method to Stimulate Cytokine Producti on and Its Use in Immunotoxicity Assessment,” Int J Environ Res Public Health, Sep. 2013, 10(9):3834-3842.
Barroga et al., “Discovery of an Intra-Articular Injection Small Molecule Inhibitor of the Wnt Pathway (SM04690) as a Potential Disease Modifying Treatment for Knee Osteoarthritis,” 2015 ACR/ARHP Annual Meeting, Abst. No. 2007, Sep. 29, 2015, retrieved on Sep. 27, 2018, URL <https://acrabstracts.org/abstract/discovery-of-an-intra-articular-injection-small-molecule-inhibitor-of-the-wnt-pathway-sm04690-as-a-potential-disease-modifying-treatment-for-knee-osteoarthritis/>, 3 pages.
Bass, “Why the difference between tendinitis and tendinosis matters,” International Journal of Therapeutic Massage and Bodywork, vol. 5, No. 1, Mar. 2012.
Bendele, “Animal Models of Arthritis: Relevance to Human Disease” Toxicol Pathol 1999 27:134-142.
Bone fractures—https://my.clevelandclinic.org/health/diseases/15241-bone-fractures—Jun. 2018, 5 pages.
Cancer definition in MedicineNet.com-2005, 1 page.
Cancer Drug Design and Discovery Neidle, Stephen, ed. (Elsevier/Academic Press, 2008), 5 pages.
Chanput et.al., “Transcription profiles of LPS-stimulated THP-1 monocytes and macrophages: a tool to study inflammation modulating effects of food-derived compounds,” Food Funct, Dec. 2010, 1(3):254-61.
clinicaltrials.gov' [online]. ClinicalTrials.gov Identifier: NCT02095548, “Phase 1, Dose Escalation Study Evaluating the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of SM04690 in Moderate to Severe Knee Osteoarthritis (OA),” Mar. 26, 2014, [retreived on Aug. 1, 2018]. Retreived from the Internet: URL<https://clinicaltrials.gov/ct2/show/NCT02095548?term=NCT02095548&rank=1>, 7 pages.
clinicaltrials.gov' [online]. ClinicalTrials.gov Identifier: NCT02536833, “A Study Evaluating the Safety, Tolerability, and Efficacy of SM04690 Injected in the Target Knee Joint of Moderately to Severely Symptomatic Osteoarthritis Subjects,” Sep. 1, 2015, [retrieved on Aug. 1, 2018]. Retrieved from the Internet: URL<https://clinicaltrials.gov/ct2/show/NCT02536833?term=NCT02536833&rank=1>, X pages.
Damia “Contemporary pre-clinical development of anticancer agents—What are the optimal preclinical models?” European Journal of Cancer 2009, 45, 2768-2781 p. 2778.
Deshmkukh et al, “Abstract: A Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from the Orthobiologic Institue (TOBI) Annual Symposium, Las Vegas, Nevada, Jun. 7, 2018, 1 page.
Deshmkukh et al, “Abstract: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from Osteoarthritis Research Society International (OARSI), Las Vegas, Nevada, Apr. 27, 2017, 2 pages.
Deshmkukh et al, “Abstract: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from the World Congress on Osteoporosis Osteoarthritis and Musculoskeletal Disease, Florence, Italy, Mar. 23, 2017, 2 pages.
Deshmkukh et al, “Abstract: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Inhibitor of the Wnt Pathway,” Abstract from Osteoarthritis Research Society International (OARSI), Liverpool, England, Apr. 26, 2018, 3 pages.
Deshmkukh et al, “Abstract: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Wnt Pathway Inhibitor,” Abstract from the Orthobiologic Institue (TOBI) Annual Symposium, Las Vegas, Nevada, Jun. 7, 2018, 2 pages.
Deshmkukh et al, “Poster: A Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Poster from the Orthobiologic Institue (TOBI) Annual Symposium, Las Vegas, Nevada, Jun. 7, 2018, 1 page.
Deshmkukh et al, “Poster: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Poster from the World Congress on Osteoporosis Osteoarthritis and Musculoskeletal Disease, Florence, Italy, Mar. 23, 2017, 1 page.
Deshmkukh et al, “Poster: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Wnt Pathway Inhibitor,” Poster from the Orthobiologic Institue (TOBI) Annual Symposium, Las Vegas, Nevada, Jun. 7, 2018, 2 pages.
Deshmkukh et al, “Presentation: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Inhibitor of the Wnt Pathway,” Presentation from Osteoarthritis Research Society International (OARSI), Liverpool, England, Apr. 26, 2018, 17 pages.
Deshmukh et al, “Abstract #EULAR-6427: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from Annual European Congress of Rheumatology (EULAR), Madrid, Spain, Jun. 14, 2017, 2 pages.
Deshmukh et al, “Abstract #THU0522: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Inhibitor of the Wnt Pathway,” Abstract from Annual European Congress of Rheumatology (EULAR), Amsterdam, Netherlands, Jun. 13-16, 2018, 2 pages.
Deshmukh et al, “Abstract: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from Orthopaedic Research Society Annual Meeting, New Orleans, Louisiana, Mar. 19, 2017, 1 page.
Deshmukh et al, “Abstract: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Abstract from Regenerative Medicine and Biology From Development to Regeneration, St. Louis, Missouri, May 4, 2017, 2 pages.
Deshmukh et al, “Abstract: Experimental tendinopathy treatment with SM04755, a topical small molecule inhibitor of the Wnt pathway,” Abstract from Orthopaedic Research Society Annual Meeting, New Orleans, Louisiana, Mar. 10, 2018, 2 pages.
Deshmukh et al, “Poster # 1459: Experimental tendinopathy treatment with SM04755, a topical small molecule inhibitor of the Wnt pathway,” Poster from Orthopaedic Research Society Annual Meeting, New Orleans, Louisiana, Mar. 19, 2018, 1 page.
Deshmukh et al, “Poster #443: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Poster from Osteoarthritis Research Society International (OARSI), Las Vegas, Nevada, Apr. 27, 2017, 1 page.
Deshmukh et al, “Poster #SAT067: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Poster from Annual European Congress of Rheumatology (EULAR), Madrid, Spain, Jun. 14, 2017, 1 page.
Deshmukh et al, “Poster #THU0522: Experimental Tendinopathy Treatment with SM04755, a Topical Small Molecule Inhibitor of the Wnt Pathway,” Poster from Annual European Congress of Rheumatology (EULAR), Amsterdam, Netherlands, Jun. 13-16, 2018, 1 page.
Deshmukh et al, “Poster: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Poster from Regenerative Medicine and Biology From Development to Regeneration, St. Louis, Missouri, May 4, 2017, 2 pages.
Deshmukh et al, “Presentation: Discovery of a Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Treatment for Tendinopathy,” Presentation from Orthopaedic Research Society Annual Meeting, New Orleans, Louisiana, Mar. 19, 2017, 19 pages.
Deshmukh et al., “A small-molecule inhibitor of the Wnt pathway (SM04690) as a potential disease modifying agent for the treatment of osteoarthritis of the knee,” Osteoarthritis and Cartilage, Jan. 2018, 26(1):18-27.
Deshmukh et al., “Abstract #1104: Discovery of a Small Molecule Inhibitor of the Wnt Pathway(SM04755) as a Potential Topical Treatment for Chronic Tendinopathy,” Abstract from 2016 ACR/ARHP Annual Meeting, Nov. 14, 2016, 3 pages.
Deshmukh et al., “Experimental Tendinopathy Topical, Small Molecule Inhibitor of the Treatment with SM04755, a Wnt Pathway,” Abstract of Oral Presentation at #1952 at the American College of Rheumatology (ACR) Conference 2018, Chicago, Illinois, Oct. 19-24, 2018, 2 pages.
Deshmukh et al., “Experimental Tendinopathy Treatment with SM04755, a Topical, Small Molecule Inhibitor of the Wnt Pathway,” Slides Present at #1952 at the American College of Rheumatology (ACR) Conference 2018, Chicago, Illinois, Oct. 19-24, 2018, 22 pages.
Deshmukh et al., “Poster #1104: Discovery of a Small Molecule Inhibitor of the Wnt Pathway(SM04755) as a Potential Topical Treatment for Chronic Tendinopathy,” Poster from 2016 ACR/ARHP Annual Meeting, Nov. 14, 2016, 3 pages.
Doumpas et al., “TCF/LEF dependent and independent transcriptional regulation of Wnt/b-catenin target genes” The EMBO Journal Nov. 13, 2018 1-14.
Enzo et al., “The Wnt/β-catenin pathway in human fibrotic-like diseases and its eligibility as a therapeutic target,” Molecular and Cellular Therapies, 2015, 3(1), 13 pages.
Forestier et al., “Prevalence of generalized osteoarthritis in a population with knee osteoarthritis,” Joint Bone Spine, May 2011, 78(3):275-278.
Gitter et al., “Characteristics of human synovial fibroblast activation by IL-1 beta and TNF alpha,” Immunology, Feb. 1989, 66(2):196-200.
Gunther et al., “Prevalence of generalised osteoarthritis in patients with advanced hip and knee osteoarthritis: the Ulm Osteoarthritis Study,” Ann. Rheum. Dis., Dec. 1998, 57(12):717-723.
Hayami et al., “Characterization of articular cartilage and subchondral bone changes in the rat anterior cruciate ligament transection and meniscectomized models of osteoarthritis,” Bone, Feb. 2006, 38(2):234-243.
Ikejima et al., “Interleukin-1 induces tumor necrosis factor (TNF) in human peripheral blood mononuclear cells in vitro and a circulating TNF-like activity in rabbits,” J Infect Dis, Jul. 1990, 162(1):215-23.
International Preliminary Report on Patentability for Application No. PCT/US2017/057536, dated Apr. 23, 2019, 8 pages.
International Preliminary Report on Patentability in International Application No. PCT/US2017/057536, dated Apr. 23, 2019, 10 pages.
Johnson, et. al. “Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials.” British Journal of Cancer 2001, 84, 1424-1431.
Lala and Orucevic, “Role of nitric oxide in tumor progression: lessons from experimental tumors,” Cancer and Metastasi Review, vol. 17, Mar. 1998, pp. 91-106.
Ledford “US cancer institute overhauls cell lines” Nature Feb. 25, 2016 vol. 530 p. 391.
McMahon et al, “VEGF receptor signaling in tumor angiogenesis,” The Oncologist, 2005, pp. 3-10.
MedlinePlus, [online] “Cancer,” [retrieved on Jul. 6, 2007]. Retrieved from the internet, URL http://www.nInn.nih.govinnedlineplus/cancer.html>.
Monner et al., “Induction of lymphokine synthesis in peripheral blood mononuclear cells with phorbol ester and calcium ionophore allows precise measurement of individual variations in capacity to produce IL 2,” Lymphokine Res. 1986;5 Suppl 1:S67-73.
Mora et al, “Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease,” Nat Rev Drug Discov. Oct. 30, 2017; 16(11): 810.
Ngkelo et. al., “LPS induced inflammatory responses in human peripheral blood mononuclear cells is mediated through NOX4 and Gia dependent PI-3 kinase signaling,” Journal of Inflammation, Dec. 2012, 9(1): 1, 7 pages.
Ocana, A. “Preclinical development of molecular targeted agents for cancer” Nat. Rev. Clin. Oncol. 2011, 8, 200-209.
Osteoarthritis, https://www.mayoclinic.org/diseases-conditions/osteoarthritis/diagnosis-treatrnent/drc-20351930—Sep. 2018, 8 pages.
Park et. al., “Optimized THP-1 differentiation is required for the detection of responses to weak stimuli,” Inflamm Res, Jan. 2007, 56(1):45-50.
Patani and Lavoie, “Bioisosterism: A Rational Approach in Drug Design,” Chem. Rev, Jul. 25, 1996, vol. 96, p. 3147-3176.
Pinedo & Slamon, “Translational Research: the role of VEGF in tumor angiogenesis,” The Oncologist, 2005, pp. 1-2.
Pritzker et al., “Osteoarthritis cartilage histopathology: grading and staging,” Osteoarthr. Cantil., Jan. 2006, 14(1):13-29.
Sperber et al., “Cytokine secretion induced lamina propria lymphocytes, and intraepithelial by superantigens in peripheral blood mononuclear cells, lymphocytes,” Clin Diagn Lab Immunol, Jul. 1995, 2(4):473-477.
Types of Brain Cancer at http://www.cancercenter.corn/brain-cancer/types-of-brain-cancer.cfrn (Mar. 12, 2013), 3 pages.
Types of Breast Cancer, published in breastcancer.org (Sep. 30, 2012), 1 page.
Yan et al., “Discovery of small molecule inhibitors of the Wnt/b-catenin signaling pathway by targeting b-catenin/Tcf4 interactions” Experimental Biology and Medicine vol. 242 Jun. 2017 1185-1197.
Yazici et al., “Abstract #: 312: Safety, Efficacy and Biomarker Outcomes of a Novel, Intra-Articular, Injectable, Wnt Inhibitor (SM04690) in the Treatment of Osteoarthritis of the Knee: Interim, Exploratory Analysis of Results from a Randomized, Double-Blind, Placebo-Controlled Phase 1 Study,” Poster, Presented at 2015 ACR/American College of Rheumatology Annual Meeting, San Francisco CA, Nov. 6-11, 2015; Arthritis Rheumatol. 2015; 67 (suppl 10): 1 page.
Yazici et al., “Abstract #: 313: Magnetic Resonance Imaging Outcomes Using an Intra-Articular Injection (SM04690) in the Treatment of Osteoarthritis of the Knee: Interim, Exploratory Analysis of Results from a Randomized, Double-Blind, Placebo-Controlled, Phase 1 Study,” Poster, Presented at 2015 ACR/American College of Rheumatology Annual Meeting, San Francisco CA, Nov. 6-11, 2015; Arthritis Rheumatol. 2015; 67 (suppl 10): 1 page.
Zhan et al., “Wnt signaling in cancer” Oncogene (2017) 36, 1461-1473.
Zheng “Small-molecule inhibitors of Wnt signaling pathway: towards novel anticancer therapeutics” Future Med. Chem. (2015) 7(18), 2485-2505.
Related Publications (1)
Number Date Country
20180185343 A1 Jul 2018 US
Provisional Applications (1)
Number Date Country
62411478 Oct 2016 US