Thiazolyl and oxazolyl urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors

Information

  • Patent Grant
  • 9981959
  • Patent Number
    9,981,959
  • Date Filed
    Tuesday, November 12, 2013
    10 years ago
  • Date Issued
    Tuesday, May 29, 2018
    6 years ago
Abstract
Compounds of Formula (I) or stereoisomers, tautomers, or pharmaceutically acceptable salts, solvates or prodrugs thereof, wherein Ring A, Ring C and X are as defined herein, are inhibitors of TrkA kinase and are useful in the treatment of diseases which can be treated with a TrkA kinase inhibitor such as pain, cancer, inflammation, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome.
Description
BACKGROUND OF THE INVENTION

The present invention relates to novel compounds, to pharmaceutical compositions comprising the compounds, to processes for making the compounds and to the use of the compounds in therapy. More particularly, it relates to thiazolyl and oxazolyl urea, thiourea, guanidine and cyanoguanidine compounds which exhibit TrkA kinase inhibition, and which are useful in the treatment of pain, cancer, inflammation/inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome.


The current treatment regimens for pain conditions utilize several classes of compounds. The opioids (such as morphine) have several drawbacks including emetic, constipatory and negative respiratory effects, as well as the potential for addictions. Non-steroidal anti-inflammatory analgesics (NSAIDs, such as COX-1 or COX-2 types) also have drawbacks including insufficient efficacy in treating severe pain. In addition, COX-1 inhibitors can cause ulcers of the mucosa. Accordingly, there is a continuing need for new and more effective treatments for the relief of pain, especially chronic pain.


Trk's are the high affinity receptor tyrosine kinases activated by a group of soluble growth factors called neurotrophins (NT). The Trk receptor family has three members: TrkA, TrkB and TrkC. Among the neurotrophins are (i) nerve growth factor (NGF) which activates TrkA, (ii) brain-derived neurotrophic factor (BDNF) and NT-4/5 which activate TrkB and (iii) NT3 which activates TrkC. Trk's are widely expressed in neuronal tissue and are implicated in the maintenance, signaling and survival of neuronal cells (Patapoutian, A. et al., Current Opinion in Neurobiology, 2001, 11, 272-280).


Inhibitors of the Trk/neurotrophin pathway have been demonstrated to be effective in numerous pre-clinical animal models of pain. For example, antagonistic NGF and TrkA antibodies such as RN-624 have been shown to be efficacious in inflammatory and neuropathic pain animal models (Woolf, C. J. et al. (1994) Neuroscience 62, 327-331; Zahn, P. K. et al. (2004) J. Pain 5, 157-163; McMahon, S. B. et al., (1995) Nat. Med. 1, 774-780; Ma, Q. P. and Woolf, C. J. (1997) NeuroReport 8, 807-810; Shelton, D. L. et al. (2005) Pain 116, 8-16; Delafoy, L. et al. (2003) Pain 105, 489-497; Lamb, K. et al. (2003) Neurogastroenterol. Motil. 15, 355-361; Jaggar, S. I. et al. (1999) Br. J. Anaesth. 83, 442-448) and neuropathic pain animal models (Ramer, M. S. and Bisby, M. A. (1999) Eur. J. Neurosci. 11, 837-846; Ro, L. S. et al. (1999); Herzberg, U. et al., Pain 79, 265-274 (1997) Neuroreport 8, 1613-1618; Theodosiou, M. et al. (1999) Pain 81, 245-255; Li, L. et al. (2003) Mol. Cell. Neurosci. 23, 232-250; Gwak, Y. S. et al. (2003) Neurosci. Lett. 336, 117-120).


It has also been shown that NGF secreted by tumor cells and tumor invading macrophages directly stimulates TrkA located on peripheral pain fibers. Using various tumor models in both mice and rats, it was demonstrated that neutralizing NGF with a monoclonal antibody inhibits cancer related pain to a degree similar or superior to the highest tolerated dose of morphine. Because TrkA kinase may serve as a mediator of NGF driven biological responses, inhibitors of TrkA and/or other Trk kinases may provide an effective treatment for chronic pain states.


Recent literature has also shown that overexpression, activation, amplification and/or mutation of Trk kinases are associated with many cancers including neuroblastoma (Brodeur, G. M., Nat. Rev. Cancer 2003, 3, 203-216), ovarian (Davidson. B., et al., Clin. Cancer Res. 2003, 9, 2248-2259), colorectal cancer (Bardelli, A., Science 2003, 300, 949), melanoma (Truzzi, F., et al., Dermato-Endocrinology 2008, 3 (1), pp. 32-36), head and neck cancer (Yilmaz, T., et al., Cancer Biology and Therapy 2010, 10 (6), pp. 644-653), gastric carcinoma (Du, J. et al., World Journal of Gastroenterology 2003, 9 (7), pp. 1431-1434), lung carcinoma (Ricci A., et al., American Journal of Respiratory Cell and Molecular Biology 25 (4), pp. 439-446), breast cancer (Jin, W., et al., Carcinogenesis 2010, 31 (11), pp. 1939-1947), Glioblastoma (Wadhwa, S., et al., Journal of Biosciences 2003, 28 (2), pp. 181-188), medulloblastoma (Gruber-Olipitz, M., et al., Journal of Proteome Research 2008, 7 (5), pp. 1932-1944), secratory breast cancer (Euthus, D. M., et al., Cancer Cell 2002, 2 (5), pp. 347-348), salivary gland cancer (Li, Y.-G., et al., Chinese Journal of Cancer Prevention and Treatment 2009, 16 (6), pp. 428-430), papillary thyroid carcinoma (Greco, A., et al., Molecular and Cellular Endocrinology 2010, 321 (1), pp. 44-49) and adult myeloid leukemia (Eguchi, M., et al., Blood 1999, 93 (4), pp. 1355-1363). In preclinical models of cancer, non-selective small molecule inhibitors of TrkA, B and C were efficacious in both inhibiting tumor growth and stopping tumor metastasis (Nakagawara, A. (2001) Cancer Letters 169:107-114; Meyer, J. et al. (2007) Leukemia, 1-10; Pierottia, M. A. and Greco A., (2006) Cancer Letters 232:90-98; Eric Adriaenssens, E., et al. Cancer Res (2008) 68:(2) 346-351).


In addition, inhibition of the neurotrophin/Trk pathway has been shown to be effective in treatment of pre-clinical models of inflammatory diseases with NGF antibodies or non-selective small molecule inhibitors of TrkA. For example, inhibition of the neurotrophin/Trk pathway has been implicated in preclinical models of inflammatory lung diseases including asthma (Freund-Michel, V; Frossard, N., Pharmacology & Therapeutics (2008) 117(1), 52-76), interstitial cystitis (Hu Vivian Y; et. al. The Journal of Urology (2005), 173(3), 1016-21), bladder pain syndrome (Liu, H.-T., et al., (2010) BJU International, 106 (11), pp. 1681-1685), inflammatory bowel diseases including ulcerative colitis and Crohn's disease (Di Mola, F. F, et. al., Gut (2000) 46(5), 670-678) and inflammatory skin diseases such as atopic dermatitis (Dou, Y.-C., et. al. Archives of Dermatological Research (2006) 298(1), 31-37), eczema and psoriasis (Raychaudhuri, S. P., et al., J. Investigative Dermatology (2004) 122(3), 812-819).


The TrkA receptor is also thought to be critical to the disease process in the infection of the parasitic infection of Trypanosoma cruzi (Chagas disease) in human hosts (de Melo-Jorge, M. et al., Cell Host & Microbe (2007) 1(4), 251-261).


Trk inhibitors may also find use in treating disease related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases. Bone metastases are a frequent complication of cancer, occurring in up to 70 percent of patients with advanced breast or prostate cancer and in approximately 15 to 30 percent of patients with carcinoma of the lung, colon, stomach, bladder, uterus, rectum, thyroid, or kidney. Osteolytic metastases can cause severe pain, pathologic fractures, life-threatening hypercalcemia, spinal cord compression, and other nerve-compression syndromes. For these reasons, bone metastasis is a serious and costly complication of cancer. Therefore, agents that can induce apoptosis of proliferating osteoblasts would be highly advantageous. Expression of TrkA receptors has been observed in the bone-forming area in mouse models of bone fracture (K. Asaumi, et al., Bone (2000) 26(6) 625-633). In addition, localization of NGF was observed in almost all bone-forming cells (K. Asaumi, et al.). Recently, it was demonstrated that a Trk inhibitor inhibits the tyrosine signaling activated by neurotrophins binding to all three of the Trk receptors in human hFOB osteoblasts (J. Pinski, et al., (2002) 62, 986-989). These data support the rationale for the use of Trk inhibitors for the treatment of bone remodeling diseases, such as bone metastases in cancer patients.


Trk inhibitors may also find use in treating diseases and disorders such as Sjogren's syndrome (Fauchais, A. L., et al., (2009) Scandinavian Journal of Rheumatology, 38(1), pp. 50-57), endometriosis (Barcena De Arellano, M. L., et al., (2011) Reproductive Sciences, 18(12), pp. 1202-1210; Barcena De Arellano, et al., (2011) Fertility and Sterility, 95(3), pp. 1123-1126; Cattaneo, A., (2010) Current Opinion in Molecular Therapeutics, 12(1), pp. 94-106), diabetic peripheral neuropathy (Kim, H. C., et al., (2009) Diabetic Medicine, 26 (12), pp. 1228-1234; Siniscalco, D., et al., (2011) Current Neuropharmacology, 9(4), pp. 523-529; Ossipov, M. H., (2011) Current Pain and Headache Reports, 15(3), pp. 185-192), and prostatitis and pelvic pain syndrome (Watanabe, T., et al., (2011) BJU International, 108(2), pp. 248-251; and Miller, L. J., et al., (2002) Urology, 59(4), pp. 603-608).


Several classes of small molecule inhibitors of Trk kinases said to be useful for treating pain or cancer are known (Expert Opin. Ther. Patents (2009) 19(3), 305-319).


SUMMARY OF THE INVENTION

It has now been found that pyrrolidinyl urea, thiourea, guanidine and cyanoguanidine compounds are inhibitors of TrkA, and useful for treating disorders and diseases such as pain, including chronic and acute pain. Compounds of the invention useful in the treatment of multiple types of pain including inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, or bone fracture. In addition, compounds of the invention are useful for treating cancer, inflammation or inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome, and diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases.


More specifically, provided herein are compounds of Formula I:




embedded image


or stereoisomers, tautomers, or pharmaceutically acceptable salts, solvates or prodrugs thereof, wherein Ring A, Ring C and X are as defined herein.


Another aspect of the present invention provides methods of treating a disease or disorder modulated by TrkA, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention or a stereoisomer, solvate or pharmaceutically acceptable salt thereof. In one embodiment, the disease and disorders include chronic and acute pain, including but not limited to inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, or bone fracture. In another embodiment, the disease and disorders include, but are not limited to, cancer, inflammation or inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome, and diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases. In one embodiment, the treatment includes treating the mammal with a compound of this invention in combination with an additional therapeutic agent.


Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention or a pharmaceutically acceptable salt thereof.


Another aspect of the present invention provides the compounds of the present invention for use in therapy.


Another aspect of the present invention provides the compounds of the present invention for use in the treatment of disease and disorders such as chronic and acute pain, including but not limited to inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, or bone fracture. Another aspect of the present invention provides the compounds of the present invention for use in the treatment of disease and disorders selected from cancer, inflammation or inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome, and diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases.


Another aspect of the present invention provides the use of a compound of this invention in the manufacture of a medicament for the treatment of disease and disorders such as chronic and acute pain including, but not limited to, inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, or bone fracture.


Another aspect of the present invention provides the use of a compound of this invention in the manufacture of a medicament for the treatment of disease and disorders selected from cancer, inflammation or inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome, and diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases.


Another aspect of the present invention provides intermediates for preparing compounds of Formula I.


Another aspect of the present invention includes methods of preparing, methods of separation, and methods of purification of the compounds of this invention.







DETAILED DESCRIPTION OF THE INVENTION

Provided herein are compounds, and pharmaceutical formulations thereof, that are useful in the treatment of diseases, conditions and/or disorders modulated by TrkA.


A representative compound of the invention (See Table B below), was found to be highly selective for TrkA over a panel of about 230 other kinases at 10 μconcentration. In addition, compounds of the invention such as those shown in Table A below, were found to be at least 1000 fold more selective for TrkA versus p38α.


One embodiment provides a compound of Formula I:




embedded image


or stereoisomers, tautomers, or pharmaceutically acceptable salts, solvates or prodrugs thereof, wherein:


X is O, S, NH or N—CN;


Ring A is




embedded image


D is O or S;


R1 is phenyl optionally substituted with one or more substituents independently selected from halogen and (1-3C)alkyl;


R2 is (1-6C)alkyl [optionally substituted with 1 to 5 fluoros] or (3-6C)cycloalkyl [optionally substituted with one or two fluoros];


Ring C is formula C-1 or C-2




embedded image


R3 is (1-6C)alkyl, hydroxy(1-6C)alkyl, Ar2, hetCyc1, (3-7C)cycloalkyl, or hetAr2;


Ar2 is phenyl optionally substituted with one or more groups independently selected from halogen and (1-6C)alkyl;


hetCyc1 is a 5-6-membered saturated or partially unsaturated heterocyclic ring having 1-2 ring heteroatoms independently selected from N and 0;


hetAr2 is a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, O and S and optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen;


R4 is H, OH, (1-6C)alkyl, monofluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, tetrafluro(2-6C)alkyl, pentafluro(2-6C)alkyl, cyano(1-6C)alkyl, hydroxy(1-6C)alkyl, dihydroxy(2-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, amino(1-6C)alkyl, aminocarbonyl(1-6C)alkyl, (1-3C)alkylsulfonamido(1-6C)alkyl, sulfamido(1-6C)alkyl, hydroxycarbonyl(1-6C)alkyl, hetAr3(1-6C)alkyl, Ar3(1-6C)alkyl, (1-6C)alkoxy, monofluoro(1-6C)alkoxy, difluoro(1-6C)alkoxy, trifluoro(1-6C)alkoxy, tetrafluoro(2-6C)alkoxy, pentafluoro(2-6C)alkoxy, cyano(1-6C)alkoxy, hydroxy(1-6C)alkoxy, dihydroxy(2-6C)alkoxy, amino(2-6C)alkoxy, hydroxyl-carbonyl(1-6C)alkoxy, hetCyc2(1-6C)alkoxy, hetAr3(1-6C)alkoxy, Ar3(1-6C)alkoxy, (1-4C alkoxy)(1-6C)alkoxy, (1-3C alkylsulfonyl)(1-6C)alkoxy, (3-6C)cycloalkyl [optionally substituted with F, OH, (1-6C alkyl), (1-6C) alkoxy, or (1-3C alkoxy)(1-6C)alkyl], hetAr4, hetAr4—O—, Ar4, hetCyc2(O)CH2—, (1-4C alkoxycarbonyl)(1-6C)alkoxy, hydroxycarbonyl(1-6C)alkoxy, aminocarbonyl(1-6C)alkoxy, hetCyc2C(═O)(1-6C)alkoxy, hydroxy(1-3C alkoxy)(1-6C)alkoxy, hydroxytrifluoro(1-6C)alkoxy, (1-3C)alkylsulfonamido(1-6C)alkoxy, (1-3C)alkylamido(1-6C)alkoxy, di(1-3C alkyl)amino-carboxy, hetCyc2C(═O)O—, hydroxydifluoro(1-6C)alkyl, (1-4C alkylcarboxy)(1-6C)alkyl, (1-6C)alkoxycarbonyl, hydroxylcarbonyl, aminocarbonyl, (1-3C alkoxy)aminocarbonyl, hetCyc3, halogen, CN, trifluoromethylsulfonyl, N-(1-3C alkyl)oxadiazolonyl, or hetAr5;


hetCyc2 is a 4-6 membered heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O and optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkylcarboxy)(1-6C)alkyl, and (1-6C)acyl;


hetCyc3 is a 4-7 membered heterocycle having 1-2 ring heteroatoms independently selected from N and O and optionally substituted with one or more substituents independently selected from F, CN, (1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, (1-6C)acyl-, (1-6C)alkylsulfonyl, trifluoromethylsulfonyl and (1-4C alkoxy)carbonyl;


hetAr3 is a 5-membered heteroaryl ring having 1-3 ring atoms independently selected from N, O and S and optionally substituted with (1-6C)alkyl;


Ar3 is phenyl optionally substituted with (1-4C)alkoxy;


hetAr4 is a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, S and O and optionally substituted with one or more substituents independently selected from (1-6C)alkyl, halogen, CN, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, difluoro(1-6C)alkyl, fluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2— (3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, (1-3C trifluoroalkoxy), fluoro(1-6C alkyl)amino, difluoro(1-6C alkyl)amino, trifluoro(1-6C alkyl)amino, and (3-4C cycloalkyl)amino;


hetAr5 is a group selected from the structures:




embedded image


where Rz is (3-4C)cycloalkyl or (1-3C)alkyl (optionally substituted with 1-3 fluoros), wherein each of said hetAr5 groups is optionally further substituted with one or more groups independently selected from F and (1-3C)alkyl optionally substituted with 1-3 fluoros;


Ar4 is phenyl optionally substituted with one or more groups independently selected from (1-6C)alkyl, halogen, CN, CF3, CF3O—, (1-6C)alkoxy, (1-6C alkyl)OC(═O)—, aminocarbonyl, (1-6C)alkylthio, hydroxy(1-6C)alkyl, (1-6C alkyl)SO2—, HOC(═O)— and (1-3C alkoxy)(1-3C alkyl)OC(═O)—;


R5 is (1-6C)alkyl, monofluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, tetrafluoro(2-6C)alkyl, pentafluoro(2-6C)alkyl, halogen, CN, (1-4C)alkoxy, hydroxy(1-4C)alkyl, (1-3C alkoxy)(1-4C)alkyl, (1-4C alkyl)OC(═O)—, (1-6C)alkylthio, (3-4C)cycloalkyl, amino, aminocarbonyl, trifluoro(1-3C alkyl)amido, or phenyl (optionally substituted with one or more groups independently selected from halogen, (1-6C)alkyl and (1-6C)alkoxy); or


R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated, partially unsaturated or unsaturated carbocyclic ring optionally substituted with one or more substituents independently selected from (1-6C)alkyl, or


R4 and R5 together with the atoms to which they are attached form 5-6 membered saturated, partially unsaturated or unsaturated heterocyclic ring having a ring heteroatom selected from N, O or S, wherein said heterocyclic ring is optionally substituted with one or two substituents independently selected from (1-6C alkyl)C(═O)O—, (1-6C)acyl, (1-6C)alkyl and oxo, and said sulfur ring atom is optionally oxidized to S(═O) or SO2;


R3a is hydrogen, halogen, (1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, phenyl optionally substituted with one or more substituents independently selected from halogen and (1-6C)alkyl, or a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, O and S and optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen;


R4a is hydrogen, (1-6C)alkyl, trifluoro(1-6C)alkyl, phenyl [optionally substituted with one or more groups independently selected from (1-6C)alkyl, halogen, CN, CF3, CF3O—, (1-6C)alkoxy, (1-6Calkyl)OC(═O)—, aminocarbonyl, (1-6C)alkylthio, hydroxy(1-6C)alkyl, (1-6C alkyl)SO2—, HOC(═O)— and (1-3C alkoxy)(1-3C alkyl)OC(═O)—], or a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, S and O and optionally substituted with 1-2 substituents independently selected from (1-6C)alkyl, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2-(3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino and (1-3C trifluoroalkoxy)(1-3C)trifluoroalkyl; and


R5a is hydrogen, halogen, (1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, phenyl optionally substituted with one or more substituents independently selected from halogen and (1-6C)alkyl, or a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, O and S and optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen.


It is to be understood that in instances where two or more radicals are used in succession to define a substituent attached to a structure, the first named radical is considered to be terminal and the last named radical is considered to be attached to the structure in question. Thus, for example, the radical “alkoxyalkyl” is attached to the structure in question by the alkyl group.


The terms “(1-6C)alkyl”, “(1-4C)alkyl” and “(1-3C)alkyl” as used herein refer to saturated linear monovalent hydrocarbon radicals of one to six carbon atoms, one to four carbon atoms, and one to three carbon atoms, respectively, or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, three to four carbon atoms, or three carbon atoms, respectively. Examples include, but are not limited to, methyl, ethyl, 1-propyl, 2-propyl, 1-butyl, 2-methyl-1-propyl, 2-butyl, 2-methyl-2-propyl, 2,2-dimethylpropyl, 1-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2-methyl-3-pentyl, 2,3-dimethyl-2-butyl, and 3,3-dimethyl-2-butyl.


“(1-4C)Alkoxy”, “(1-3C)alkoxy”, “(1-6C)alkoxy” and “(2-6C)alkoxy” refer to an —OR radical where R is (1-4C)alkyl, (1-3C)alkyl, (1-6C)alkyl, or (2-6C)alkyl, respectively, as defined above. Examples include methoxy, ethoxy, and the like.


“(1-6C)acyl” means a RC(═O)— radical where R is a linear saturated monovalent hydrocarbon radical of one to five carbon atoms or a branched saturated monovalent hydrocarbon radical of three to five carbon atoms, e.g., methylcarbonyl, and the like.


“(1-3C Alkoxy)(1-6C)alkyl” and “(1-3C alkoxy)(1-4C)alkyl” mean a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or one to four carbon atoms, or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms or three to four carbon atoms, respectively, wherein one of the carbon atoms is substituted with one (1-3C)alkoxy group as defined herein.


“(1-3C Alkoxy)(1-6C)alkoxy” means a (1-6C)alkoxy group as defined herein, wherein one of the carbon atoms is substituted with a (1-3C)alkoxy group as defined herein. Examples include methoxymethoxy, methoxyethoxy, and the like.


“(1-3C Alkoxy)aminocarbonyl” means a (1-3C alkyl)-O—NH—C(═O)— group.


“(1-6C)Alkoxycarbonyl” means a (1-6C)—O—C(═O)— group.


“Amino” means a —NRR′ group where R and R′ are independently selected from hydrogen or (1-3C)alkyl as defined herein. Examples include H2N—, CH3NH—, (CH3)2N, and the like.


“Amino(1-6C)alkyl” means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, wherein one of the carbon atoms is substituted with one —NRR′ group where R and R′ are independently selected from hydrogen or (1-3C)alkyl as defined herein. Examples include aminomethyl, methylaminoethyl, 2-ethylamino-2-methylethyl, and the like.


“Amino(2-6C)alkoxy” means a (2-6C)alkoxy group as defined herein, wherein one of the carbon atoms is substituted with one —NRR′ group where R and R′ are independently selected from hydrogen or (1-3C)alkyl as defined herein.


“Aminocarbonyl” means a RR′NCO— radical where R and R′ are independently hydrogen or (1-6C)alkyl as defined herein. Examples include H2NCO—, dimethylaminocarbonyl, and the like.


“Aminocarbonyl(1-6C)alkyl” means a linear saturated hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbons wherein one of the carbon atoms is substituted with one aminocarbonyl group as defined herein, e.g., 2-aminocarbonylethyl, 1-, 2-, or 3-dimethylaminocarbonylpropyl, and the like.


“Aminocarbonyl(1-6C)alkoxy” means a (1-6C)alkoxy as defined herein, wherein one of the carbon atoms is substituted with one aminocarbonyl group as defined herein.


“(1-3C)Alkylamido(1-6C)alkoxy” means a (1-6C)alkoxy as defined herein, wherein one of the carbon atoms is substituted with one alkylamido group, i.e., substituted with a (1-3C)C(═O)NH— group.


“(1-3C)Alkylsulfonamido” means a (1-3C)alkylSO2NH— radical where (1-3C)alkyl is as defined herein


“(1-3C Alkylsulfonamido)(1-6C)alkyl” means a linear saturated hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbons substituted with one (1-3C)alkylsulfonamido group as defined herein.


“(1-3C)Alkylsulfonamido(1-6C)alkoxy” means a (1-6C)alkoxy group as defined herein wherein one of the carbon atoms is substituted with one (1-3C)alkylsulfonamido group as defined herein.


“Hydroxycarbonyl” means HOC(═O)—.


“Cyano(1-6C)alkyl” means a linear saturated hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbons substituted with a cyano (CN) group.


“(3-6C)Cycloalkyl” means a cyclic saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.


“Dihydroxy(2-6C)alkyl” means a linear saturated hydrocarbon radical of two to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbons substituted with two hydroxy (OH) groups, provided that two hydroxy groups are not both on the same carbon atom.


“Dihydroxy(2-6C)alkoxy” means a (2-6C)alkoxy group as defined herein, wherein two of the carbon atoms are substituted with a hydroxy group.


“Halogen” as used herein means F, Cl, Br or I.


“Heterocycle” refers to a saturated or partially unsaturated ring system having one or more ring heteroatoms as recited for the specific heterocyclic group, wherein the heterocycle is optionally substituted with substituents as defined for that particular heterocyclic group.


“Heteroaryl” refers to a 5-6 membered unsaturated ring system having one or more ring heteroatoms as recited for the specific heteroaryl group, wherein the heteroaryl is optionally substituted with substituents as defined for that particular heteroaryl group.


“hetCyc2C(═O)(1-6C)alkoxy” means a (1-6C)alkoxy as defined herein, wherein one of the carbon atoms is substituted with a hetCyc2C(═O) group, wherein hetCyc2 is as defined herein.


“Hydroxy(1-6C)alkyl” and “hydroxy(1-4C)alkyl” means a linear saturated hydrocarbon radical of one to six carbon atoms or one to four carbon atoms, respectively, or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms or three to four carbon atoms, respectively, wherein one of the carbon atoms is substituted with a hydroxy (OH) group.


“Hydroxy(1-6C)alkoxy” means a (1-6C)alkoxy group as defined herein, wherein one of the carbon atoms is substituted with a hydroxy group.


“Hydroxy(1-3C alkoxy)(1-6C)alkoxy” means a (1-3C alkoxy)(1-6C)alkoxy as defined herein, wherein one of the carbon atoms is substituted with a hydroxy group.


“Hydroxydifluoro(1-6C)alkyl” means a difluoro(1-6C)alkyl group as defined herein, wherein one of the carbon atoms is substituted with a hydroxy group.


“Hydroxytrifluoro(1-6C)alkoxy” means a trifluoro(1-6C)alkoxy group as defined herein, wherein one of the carbon atoms is substituted with a hydroxy group.


“Monofluoro(1-6C)alkyl”, “difluoro(1-6C)alkyl” and “trifluoro(1-6C)alkyl” refer to a (1-6C)alkyl group as defined herein wherein one to three hydrogen atoms, respectively, is replaced by a fluoro group.


“Tetrafluoro(2-6C)alkyl” and “pentafluoro(2-6C)alkyl” refer to a linear saturated monovalent hydrocarbon radical of two to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms wherein four to five hydrogen atoms, respectively, is replaced by a fluoro group.


“Trifluoro(1-3C alkyl)amido” means a (1-3C alkyl)C(═O)NH— group wherein one of the carbons is substituted with three fluoros.


“Trifluoro(1-6C)alkoxy” means a (1-6C)alkoxy group as defined herein, wherein one of the carbon atoms is substituted with three fluoros.


“Sulfamido(1-6C)alkyl” means a linear saturated hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbons substituted with one sulfamido (H2NSO2NH—) group.


It should be noted that compounds of the invention may contain groups that may exist in tautomeric forms, such as heteroatom substituted heteroaryl or heterocyclic groups and the like, which are illustrated in the following general and specific examples:




embedded image


where G′=O, S, or NR, and though one form is named, described, displayed and/or claimed herein, all the tautomeric forms are intended to be inherently included in such name, description, display and/or claim.


In one embodiment of Formula I, D is S.


In one embodiment of Formula I, D is O.


In one embodiment of Formula I, X is O.


In one embodiment, X is S.


In one embodiment, X is NH.


In one embodiment, X is N—CN.


In one embodiment of Formula I, R1 is phenyl optionally substituted with one or more substituents independently selected from halogen and (1-3C)alkyl. In one embodiment of Formula I, R1 is phenyl.


In one embodiment of Formula I, R2 is (1-6C)alkyl [optionally substituted with 1 to 5 fluoros] or (3-6C)cycloalkyl [optionally substituted with one or two fluoros].


In one embodiment of Formula I, R2 is (1-6C)alkyl optionally substituted with 1 to 5 fluoros. In one embodiment, R2 is methyl, isopropyl, trifluoromethyl or 2,2,2-trifluoroethyl. In one embodiment, R2 is methyl or trifluoromethyl.


In one embodiment of Formula I, R2 is (3-6C)cycloalkyl optionally substituted with one or two fluoros. In one embodiment, R2 is cyclopropyl, 2,2-difluorocyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.


In one embodiment, Ring C is formula C-1:




embedded image


where R3, R4 and R5 are as defined for Formula I.


In one embodiment, R3 is (1-6C)alkyl. In one embodiment, R3 is methyl or ethyl.


In one embodiment, R3 is hydroxy(1-6C)alkyl. An example of R3 is 2-hydroxyethyl.


In one embodiment, R3 is Ar2, where Ar2 is phenyl optionally substituted with one or more groups independently selected from halogen and (1-6C)alkyl.


In one embodiment, R3 when represented by Ar2 is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 3-chlorophenyl, 3-chloro-4-fluorophenyl or 3-chloro-2-fluorophenyl. In one embodiment, R3 when represented by Ar2 is phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-methylphenyl, 3-methylphenyl or 4-methylphenyl. In one embodiment, R3 is phenyl.


In one embodiment, R3 is hetCyc1, where hetCyc1 is a 5-6-membered saturated or partially unsaturated heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O. In one embodiment, R3 is a pyrrolidinyl, tetrahydrofuranyl, imidazolidinyl, piperidinyl, piperazinyl, tetrahydropyranyl, or morpholinyl ring. In one embodiment, R3 is tetrahydro-2H-pyran-4-yl.


In one embodiment, R3 is (3-7C)cycloalkyl. In one embodiment R3 is cyclohexyl.


In one embodiment, R3 is hetAr2, where hetAr2 is 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, O and S and optionally substituted with one or more substituents independently selected from (1-6C)alkyl and halogen. In one embodiment, R3 is thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, pyridyl, pyrimidyl, pyrazinyl, or pyridazinyl optionally substituted with one or more substituents independently selected from (1-6C)alkyl and halogen. In one embodiment, R3 is pyrazolyl, pyridyl or pyridazinyl optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen. In one embodiment, R3 is pyrazolyl, pyridyl or pyridazinyl optionally substituted with (1-6C)alkyl or halogen. In one embodiment, R3 when represented by hetAr2 is 1-methyl-1H-pyrazol-4-yl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, pyridazinyl or 3-chloropyrid-5-yl.


In one embodiment, R3 is selected from Ar2 and hetAr2.


In one embodiment, R3 is Ar2. In one embodiment, R3 is phenyl.


In one embodiment, R4 is OH.


In one embodiment, R4 is (1-6C)alkyl. In one embodiment, R4 is methyl, ethyl, isopropyl or tert-butyl.


In one embodiment, R4 is monofluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, tetrafluoro(2-6C)alkyl or pentafluoro(2-6C)alkyl. In one embodiment, R4 is fluoromethyl, 2-fluoroethyl, difluoromethyl and 2,2-difluoroethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, 2,2,3,3-tetrafluoropropyl or 2,2,3,3,3-pentafluoropropyl


In one embodiment, R4 is trifluoro(1-6C)alkyl. In one embodiment, R4 is CF3.


In one embodiment, R4 is cyano(1-6C)alkyl. In one embodiment, R4 is cyanomethyl or 2-cyanopropan-2-yl.


In one embodiment, R4 is hydroxy(1-6C)alkyl. In one embodiment, R4 is hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 2-hydroxy-2-methylpropyl or 1-hydroxy-2-methylpropan-2-yl.


In one embodiment, R4 is dihydroxy(2-6C)alkyl. In one embodiment, R4 is 2,3-dihydroxypropyl.


In one embodiment, R4 is (1-3C alkoxy)(1-6C)alkyl. In one embodiment, R4 is methoxymethyl, 2-methoxyethyl or 3-methoxypropyl.


In one embodiment, R4 is amino(1-6C)alkyl. In one embodiment, R4 is aminomethyl, 2-aminoethyl or 3-aminopropyl.


In one embodiment, R4 is aminocarbonyl(1-6C)alkyl. In one embodiment, R4 is aminocarbonylmethyl and 2-(aminocarbonyl)ethyl.


In one embodiment, R4 is (1-3C)alkylsulfonamido(1-6C)alkyl. In one embodiment, R4 is CH3SO2NHCH2— or CH3SO2NHCH2CH2—.


In one embodiment, R4 is hydroxycarbonyl(1-6C)alkyl. In one embodiment, R4 is HOC(═O)CH2— and HOC(═O)CH2CH2—.


In one embodiment, R4 is hetAr3(1-6C)alkyl, where hetAr3 is a 5-membered heteroaryl ring having 1-3 ring atoms independently selected from N, S and O and optionally substituted with (1-6C)alkyl. In one embodiment, hetAr3 is a thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl or oxadiazolyl ring optionally substituted with (1-6C)alkyl. In one embodiment, R4 when represented by hetAr3(1-6C)alkyl is (1-methyl-1H-1,2,4-triazol-3-yl)methyl or (5-methyl-1,3,4-oxadiazol-2-yl)methyl.


In one embodiment, R4 is Ar3(1-6C)alkyl, where phenyl optionally substituted with (1-4C)alkoxy or hydroxy(1-4C)alkyl. In one embodiment, Ar3(1-6C)alkyl is benzyl.


In one embodiment, R4 is (1-6C)alkoxy. Examples include methoxy and ethoxy.


In one embodiment, R4 is monofluoro(1-6C)alkoxy, difluoro(1-6C)alkoxy trifluoro(1-6C)alkoxy, tetrafluoro(2-6C)alkoxy or pentafluoro(2-6C)alkoxy. In one embodiment, R4 is fluoromethoxy, 2-fluoroethoxy, 2,2-difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy or 2,2-difluoroethoxy. In one embodiment, R4 is 2-fluoroethoxy.


In one embodiment, R4 is cyano(1-6C)alkoxy. In one embodiment, R4 is cyanomethoxy or 2-cyanoethoxy.


In one embodiment, R4 is hydroxy(1-6C)alkoxy. In one embodiment, R4 is 2-hydroxy-2-methylpropoxy, 2-hydroxyethoxy, 2-hydroxypropoxy, 2-hydroxy-2-methylpropoxy or 2-hydroxybutoxy.


In one embodiment, R4 is dihydroxy(2-6C)alkoxy. In one embodiment, R4 is 2,3-dihydroxypropoxy or 3-hydroxy-2-(hydroxymethyl)propoxy.


In one embodiment, R4 is amino(2-6C)alkoxy. In one embodiment, R4 is H2NCH2CH2O—.


In one embodiment, R4 is hetCyc2(1-6C)alkoxy, where hetCyc2 is a 4-6 membered heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O, wherein hetCyc2 is optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl, and (1-6C)acyl. In one embodiment, hetCyc2 is oxetaynyl, tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or and 1,3-dioxolanyl optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, R4 when represented by hetCyc2(1-6C)alkoxy is oxetan-2-ylmethoxy, 2-(oxetan-2-yl)propoxy, 2-morpholinoethoxy, piperazinylethyoxy or piperidinylethoxy optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, R4 is represented by the structures:




embedded image


In one embodiment, R4 is hetAr3(1-6C)alkoxy, where hetAr3 is a 5-membered heteroaryl ring having 1-3 ring atoms independently selected from N, S and O and optionally substituted with (1-6C)alkyl. In one embodiment, hetAr3 is a thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl or oxadiazolyl ring optionally substituted with (1-6C)alkyl. In one embodiment, hetAr3 is triazolyl or oxadiazolyl ring optionally substituted with a (1-6C)alkyl group such as a methyl group. In one embodiment, R4 when represented by hetAr3(1-6C)alkoxy is (1-methyl-1H-1,2,4-triazol-3-yl)methoxy or (5-methyl-1,3,4-oxadiazol-2-yl)methoxy, which can be represented by the structures:




embedded image


In one embodiment, R4 is Ar3(1-6C)alkoxy, where Ar3 is phenyl optionally substituted with (1-4C)alkoxy. In one embodiment, R4 is phenylmethoxy or (4-methoxyphenyl)methoxy having the structures:




embedded image


In one embodiment, R4 is (1-4C alkoxy)(1-6C)alkoxy. In one embodiment, R4 is (2-methoxy)ethoxy having the structure:




embedded image


In one embodiment, R4 is (1-3Calkylsulfonyl)(1-6C)alkoxy. In one embodiment, R4 is (2-methylsulfonyl)ethoxy having the structure:




embedded image


In one embodiment, R4 is (3-6C)cycloalkyl optionally substituted with F, OH, (1-6C alkyl), (1-6C)alkoxy or (1-3C alkoxy)(1-6C)alkyl. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-hydroxycyclobutyl. In one embodiment, R4 is cyclopropyl or 2-hydroxycyclobutyl. In one embodiment, R4 is cyclopropyl.


In one embodiment, R4 is hetAr4, where hetAr4 is a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, S and O and optionally substituted with one or more substituents independently selected from (1-6C)alkyl, halogen, CN, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, difluoro(1-6C)alkyl, fluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2— (3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, (1-3C trifluoroalkoxy), fluoro(1-6C alkyl)amino, difluoro(1-6C alkyl)amino, trifluoro(1-6C alkyl)amino, and (3-4C cycloalkyl)amino.


In one embodiment, R4 is hetAr4 where hetAr4 is pyridyl, pyrimidinyl pyridazinyl, pyrazolyl, imidazolyl, thienyl, 1,2,4-triazolyl, 1,2,3-triazolyl, thiazolyl, oxazolyl, 1,3,4-oxadiazolyl, or 1,2,4-oxadiazolyl optionally substituted with one or more substituents independently selected from (1-6C)alkyl, halogen, CN, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, difluoro(1-6C)alkyl, fluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2-(3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, (1-3C trifluoroalkoxy), fluoro(1-6C alkyl)amino, difluoro(1-6C alkyl)amino, trifluoro(1-6C alkyl)amino, and (3-4C cycloalkyl)amino.


In one embodiment, R4 is hetAr4 where hetAr4 is pyridyl, pyrimidinyl pyridazinyl, pyrazolyl, imidazolyl, thienyl, 1,2,4-triazolyl, 1,2,3-triazolyl, thiazolyl, oxazolyl, 1,3,4-oxadiazolyl, or 1,2,4-oxadiazolyl optionally substituted with one or more substituents independently selected from (1-6C)alkyl, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2— (3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, (1-3C trifluoroalkoxy)(1-3C)trifluoroalkyl and cyclopropylNH—.


In one embodiment, R4 is hetAr4, where hetAr4 is pyridyl, pyrimidinyl pyridazinyl, pyrazolyl, imidazolyl, thionyl, 1,2,4-triazolyl, 1,2,3-triazolyl, thiazolyl, oxazolyl, 1,3,4-oxadiazolyl, or 1,2,4-oxadiazolyl optionally substituted with 1-3 substituents independently selected from fluoro, methyl, ethyl, isopropyl, cyclopropylmethyl, cyclopropyl, trifluoromethyl, 2,2,2-trifluoroethyl, methoxy, ethoxy, CN, H2N—, (CH3)2N—, 2-hydroxyethyl, 2-methoxyethyl, 1-(2,2,2-trifluoroethoxy)-2,2,2-trifluoroethyl, cyclopropylcarbonyl, methylsulfonyl and cyclopropylNH—.


In one embodiment, R4 is hetAr4, where hetAr4 is pyrazolyl optionally substituted with one or more groups independently selected from (1-6C)alkyl.


In one embodiment, R4 when represented by hetAr4 is selected from the structures:




embedded image


embedded image


embedded image


In one embodiment, R4 is hetAr4—O—. In one embodiment, R4 is the structure:




embedded image


In one embodiment, R4 is Ar4, where Ar4 is phenyl optionally substituted with one or more groups independently selected from (1-6C)alkyl, halogen, CN, CF3, CF3O—, (1-6C)alkoxy, (1-6Calkyl)OC(═O)—, aminocarbonyl, (1-6C)alkylthio, hydroxy(1-6C)alkyl, (1-6C alkyl)SO2—, HOC(═O)— and (1-3C alkoxy)(1-3C alkyl)OC(═O)—. In one embodiment, Ar4 is phenyl optionally substituted with one or more groups independently selected from methyl, F, Cl, CN, methoxy, CH3OC(═O)—, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl, methylthio, CH3SO2—, HOC(═O)— and CH3OCH2CH2OC(═O)—. In one embodiment, Ar4 is phenyl optionally substituted with one or two of said substituents. In one embodiment, Ar4 is selected from the structures:




embedded image


In one embodiment, R4 is hetCyc2(O)CH2, where hetCyc2 is a 4-6 membered heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O, wherein hetCyc2 is optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl, and (1-6C)acyl. Examples of hetCyc2 include oxetaynyl, tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, and 1,3-dioxolanyl rings optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, R4 when represented by hetCyc2(O)CH2 is selected from the structures:




embedded image


In one embodiment, R4 is (1-4C alkoxycarbonyl)(1-6C)alkoxy. In one embodiment, R4 is methoxycarbonyl(1-6C)alkoxy or ethylcarbonyl(1-6C)alkoxy. A particular example is ethoxycarbonylmethoxy.


In one embodiment, R4 is hydroxycarbonyl(1-6C)alkoxy. In one embodiment, R4 is hydroxycarbonylmethoxy.


In one embodiment, R4 is aminocarbonyl(1-6C)alkoxy. In one embodiment, R4 is H2NC(═O)(1-6C)alkoxy, (1-6C alkyl)NHC(═O)(1-6C)alkoxy, or di(1-6Calkyl)NC(═O)(1-6C)alkoxy. In one embodiment, R4 is H2NC(═O)CH2O—, H2NC(═O)CH2CH2O—, or CH3CH2NC(═O)CH2O—.


In one embodiment, R4 is hetCyc2C(═O)(1-6C)alkoxy, where hetCyc2 is a 4-6 membered heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O and optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl, and (1-6C)acyl. In one embodiment, hetCyc2 is oxetaynyl, tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or 1,3-dioxolanyl optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, hetCyc2 is morpholinyl. In one embodiment, R4 when represented by hetCyc2C(═O)(1-6C)alkoxy is the structure:




embedded image


In one embodiment, R4 is hydroxy(1-3C alkoxy)(1-6C)alkoxy. In one embodiment, R4 is 2-hydroxy-3-methoxypropoxy, having the structure:




embedded image


In one embodiment, R4 is hydroxytrifluoro(1-6C)alkoxy. In one embodiment, R4 is 3,3,3-difluoro-2-hydroxypropoxy having the structure:




embedded image


In one embodiment, R4 is (1-3C)alkylsulfonamido(1-6C)alkoxy. In one embodiment, R4 is methanesulfonamido(1-6C)alkoxy. In one embodiment, R4 is 2-methanesulfonamidoethoxy having the structure:




embedded image


In one embodiment, R4 is (1-3C)alkylamido(1-6C)alkoxy. In one embodiment, R4 is 2-(methylamido)ethoxy having the structure:




embedded image


In one embodiment, R4 is di(1-3C alkyl)aminocarboxy. In one embodiment, R4 is dimethylaminocarboxy having the structure:




embedded image


In one embodiment, R4 is hetCyc2C(═O)O—, where hetCyc2 is a 4-6 membered heterocyclic ring having 1-2 ring heteroatoms independently selected from N and O and optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, hetCyc2 is oxetaynyl, tetrahydrofuranyl, tetrahydropyranyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or 1,3-dioxolanyl optionally substituted with 1-2 groups independently selected from (1-6C)alkyl, (1-4C alkoxy)carbonyl and (1-6C)acyl. In one embodiment, hetCyc2 is morpholinyl. In one embodiment, R4 when represented by hetCyc2C(═O)O— is the structure:




embedded image


In one embodiment, R4 is hydroxydifluoro(1-6C)alkyl. In one embodiment, R4 is 2,2-difluoro-2-hydroxyethyl.


In one embodiment, R4 is (1-4C alkylcarboxy)(1-6C)alkyl. In one embodiment, R4 is methylcarboxy(1-6C)alkyl. In one embodiment, R4 is 2-(methylcarboxy)ethyl.


In one embodiment, R4 is (1-6C)alkoxycarbonyl. In one embodiment, R4 is methoxycarbonyl or ethoxycarbonyl.


In one embodiment, R4 is hydroxycarbonyl.


In one embodiment, R4 is aminocarbonyl, that is, a RR′NCO— radical where R and R′ are independently hydrogen or (1-6C)alkyl as defined herein. In one embodiment, R4 is aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl, ethylcarbonyl or isopropylaminocarbonyl.


In one embodiment, R4 is (1-3C alkoxy)aminocarbonyl. In one embodiment, R4 is methoxyaminocarbonyl.


In one embodiment, R4 is hetCyc3, where is a 4-7 membered heterocycle having 1-2 ring heteroatoms independently selected from N and O and optionally substituted with one or more substituents independently selected from F, CN, CF3, (1-6C)alkyl, hydroxy(1-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, (1-6C)acyl-, (1-6C)alkylsulfonyl, trifluoromethylsulfonyl and (1-4C alkoxy)carbonyl. In one embodiment, hetCyc3 is tetrahydropyranyl, piperidinyl, pyrrolidinyl or azetidinyl optionally substituted with one or more substituents independently selected from F, CN, (1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, (1-6C)acyl-, (1-6C)alkylsulfonyl, trifluoromethylsulfonyl and (1-4C alkoxy)carbonyl. In one embodiment, hetCyc3 is optionally substituted with one or two of said substituents. In one embodiment, hetCyc3 is tetrahydropyranyl, piperidinyl, pyrrolidinyl or azetidinyl optionally substituted with CN, Me, CH3C(═O)—, MeSO2—, or CF3SO2—. In one embodiment, R4 when represented by hetCyc3 is selected from the structures:




embedded image


In one embodiment, R4 is halogen. In one embodiment, R4 is Br.


In one embodiment, R4 is CN.


In one embodiment, R4 is trifluoromethylsulfonyl.


In one embodiment, R4 is hetAr5, where hetAr5 is a group selected from the structures:




embedded image


where Rz is (3-4C)cycloalkyl or (1-3C)alkyl (optionally substituted with 1-3 fluoros), wherein each of said hetAr5 groups is optionally further substituted with one or more groups independently selected from F and (1-3C)alkyl optionally substituted with 1-3 fluoros.


In one embodiment, R4 when represented by hetAr5 is selected from the structures:




embedded image


In one embodiment, R4 is N-(1-3C alkyl)oxadiazolonyl. In one embodiment, R4 is represented by the structures:




embedded image


In one embodiment, R4 is selected from H, (1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C)alkyl, cyano(1-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, monofluoro(1-6C)alkoxy, cyano(1-6C)alkoxy, hydroxy(1-6C)alkoxy, dihydroxy(2-6C)alkoxy, hetCyc2(1-6C)alkoxy, Ar3(1-6C)alkoxy, (1-4C alkoxy)(1-6C)alkoxy, (1-3C alkylsulfonyl)(1-6C)alkoxy, (3-6C)cycloalkyl, hetAr4, hetAr4—O—, Ar4, and hetAr5.


In one embodiment, R4 is hetAr4, Ar4, or hetAr5.


In one embodiment, R4 is hetAr4 or hetAr5.


In one embodiment, R5 is (1-6C)alkyl. In one embodiment, R5 is methyl, ethyl, propyl, isopropyl or butyl.


In one embodiment, R5 is monofluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, tetrafluro(2-6C)alkyl or pentafluro(2-6C)alkyl. In one embodiment, R5 is fluoromethyl, 2-fluoroethyl, difluoromethyl, 2,2-difluoroethyl, 1,3-difluoroprop-2-yl, trifluoromethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, 1,1,2,2-tetrafluoropropane or 2,2,3,3,3-pentafluoropropyl.


In one embodiment, R5 is halogen. In one embodiment, R5 is F. In one embodiment, R5 is Cl. In one embodiment, R5 is Br.


In one embodiment, R5 is CN.


In one embodiment, R5 is (1-4C)alkoxy. In one embodiment, R5 is methoxy or ethoxy.


In one embodiment, R5 is hydroxy(1-4C)alkyl. In one embodiment, R5 is hydroxymethyl or 3-hydroxypropyl.


In one embodiment, R5 is (1-4C alkyl)OC(═O)—. In one embodiment, R5 is CH3CH2OC(═O)—.


In one embodiment, R5 is (1-6C)alkylthio. In one embodiment, R5 is methylthio (MeS—).


In one embodiment, R5 is phenyl optionally substituted with one or more groups independently selected from halogen, (1-6C)alkyl and (1-6C)alkoxy. In one embodiment, R5 is phenyl optionally substituted with one or more groups independently selected from F, Cl, methyl, ethyl, methoxy and ethoxy. In one embodiment, R5 is phenyl.


In one embodiment, R5 is (3-4C)cycloalkyl. In one embodiment, R5 is cyclopropyl. In one embodiment, R5 is cyclobutyl.


In one embodiment, R5 is amino. In one embodiment, R5 is NH2.


In one embodiment, R5 is aminocarbonyl. In one embodiment, R5 is H2NC(═O)—.


In one embodiment, R5 is trifluoro(1-3C alkyl)amido. In one embodiment, R5 is CF3C(═O)NH—.


In one embodiment, R5 is halogen, CN, (1-6C)alkyl, (1-4C)alkoxy, hydroxy(1-4C)alkyl, or phenyl optionally substituted with one or more groups independently selected from halogen, (1-6C)alkyl and (1-6C)alkoxy.


In one embodiment, R5 is selected from halogen, and (1-6C)alkyl.


In one embodiment, R5 is selected from methyl, Cl and Br.


In one embodiment of Formula I, R4 is selected from H, (1-6C)alkyl, trifluoro(1-6C)alkyl, cyano(1-6C)alkyl, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, cyano(1-6C)alkoxy, hydroxy(1-6C)alkoxy, (1-4C alkoxy)(1-6C)alkoxy, (3-6C)cycloalkyl, hetAr4, Ar4, and hetAr5; and R5 is selected from halogen, CN, (1-6C)alkyl, (1-4C)alkoxy, hydroxy(1-4C)alkyl, (1-6C)alkylthio, and phenyl optionally substituted with one or more groups independently selected from halogen, (1-6C)alkyl and (1-6C)alkoxy.


In one embodiment of Formula I, R4 is selected from hetAr4, Ar4, and hetAr5; and R5 is selected from (1-6C)alkyl.


In one embodiment of Formula I, R4 is selected from hetAr4 and hetAr5; and R5 is selected from (1-6C)alkyl.


In one embodiment of Formula I, R4 is hetAr4 and R5 is selected from (1-6C)alkyl.


In one embodiment of Formula I, R4 is pyrazolyl optionally substituted with one or more substituents independently selected from (1-6C)alkyl; and R5 is selected from (1-6C)alkyl.


In one embodiment of Formula I, R4 is hetAr5; and R5 is selected from (1-6C)alkyl.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated, partially unsaturated or unsaturated carbocyclic ring optionally substituted with one or more substituents independently selected from (1-6C)alkyl; or R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated, partially unsaturated or unsaturated heterocyclic ring having a ring heteroatom selected from N, O or S, wherein said heterocyclic ring is optionally substituted with one or two substituents independently selected from (1-6C alkyl)C(═O)O—, (1-6C)acyl, (1-6C)alkyl and oxo, and said sulfur ring atom is optionally oxidized to S(═O) or SO2.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated, partially unsaturated or unsaturated carbocyclic ring optionally substituted with one or more substituents independently selected from (1-6C)alkyl.


In one embodiment, Ring C when R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated or unsaturated carbocyclic ring is selected from the structures:




embedded image


where R3 is as defined for Formula I.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated carbocyclic ring optionally substituted with one or more substituents independently selected from (1-6C)alkyl, or R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated heterocyclic ring having a ring heteroatom selected from N, O or S, wherein said ring nitrogen atom is optionally substituted with (1-6C alkyl)C(═O)O— or (1-6C)acyl, and said sulfur ring atom is optionally oxidized to S(═O) or SO2.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated carbocyclic ring optionally substituted with one or more substituents independently selected from (1-6C)alkyl. In one embodiment, Ring C when R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated carbocyclic ring is selected from the structures:




embedded image


where R3 is as defined for Formula I.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated, partially unsaturated or unsaturated heterocyclic ring having a ring heteroatom selected from N, O or S, wherein said ring N atom is optionally substituted with (1-6C alkyl)C(═O)O—, (1-6C alkyl)C(═O)—, (1-6C)alkyl or oxo, and said S ring atom is optionally oxidized to S(═O) or SO2. In one embodiment, Ring C when R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated heterocyclic ring is selected from the structures:




embedded image


where R3 is as defined for Formula I.


In one embodiment, R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated heterocyclic ring having a ring heteroatom selected from N, O or S, wherein said ring N atom is optionally substituted with (1-6C alkyl)C(═O)O— or (1-6C alkyl)C(═O)—, and said S ring atom is optionally oxidized to S(═O) or SO2. In one embodiment, Ring C when R4 and R5 together with the atoms to which they are attached form a 5-6 membered saturated heterocyclic ring is selected from the structures:




embedded image


where R3 is as defined for Formula I.


In one embodiment, Ring C is formula C-2




embedded image


where R3a, R4a and R5a are as defined for Formula I.


In one embodiment, R3a is hydrogen.


In one embodiment, R3a is halogen.


In one embodiment, R3a is (1-6C)alkyl. In one embodiment, R3a is methyl.


In one embodiment, R3a is trifluoro(1-6C)alkyl. In one embodiment, R3a is CF3.


In one embodiment, R3a is (3-6C)cycloalkyl. In one embodiment, R3a is cyclopropyl.


In one embodiment, R3a is phenyl optionally substituted with one or more substituents independently selected from halogen and (1-6C)alkyl. In one embodiment, R3a is phenyl, fluorophenyl or methylphenyl, for example include phenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 3-chlorophenyl, 3-chloro-4-fluorophenyl or 3-chloro-2-fluorophenyl. In one embodiment, R3a is phenyl.


In one embodiment, R3a is a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, O and S and optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen. In one embodiment, R3a is a thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, pyridyl, pyrimidyl, pyrazinyl, or pyridazinyl ring optionally substituted with (1-6C)alkyl or halogen. In one embodiment, R3a is pyrazolyl, pyridyl or pyridazinyl optionally substituted with one or more groups independently selected from (1-6C)alkyl and halogen. In one embodiment, R3a is pyrazolyl, pyridyl or pyridazinyl optionally substituted with (1-6C)alkyl or halogen.


In one embodiment, R4a is hydrogen.


In one embodiment, R4a is (1-6C)alkyl. In one embodiment, R4a is methyl, ethyl or isopropyl.


In one embodiment, R4a is trifluoro(1-6C)alkyl. In one embodiment, R4a is 2,2,2-trifluoroethyl.


In one embodiment, R4a is phenyl optionally substituted with one or more groups independently selected from (1-6C)alkyl, halogen, CN, CF3, CF3O—, (1-6C)alkoxy, (1-6Calkyl)OC(═O)—, aminocarbonyl, (1-6C)alkylthio, hydroxy(1-6C)alkyl, (1-6C alkyl)SO2—, HOC(═O)— and (1-3C alkoxy)(1-3C alkyl)OC(═O)—. In one embodiment, R4a is phenyl optionally substituted with one or more groups independently selected from methyl, F, Cl, CN, methoxy, CH3OC(═O)—, aminocarbonyl, methylaminocarbonyl, dimethylaminocarbonyl, methylthio, CH3SO2—, HOC(═O)— or CH3OCH2CH2OC(═O)—. In certain embodiments, R4a is phenyl optionally substituted with one or two of said substituents. In one embodiment, R4a is phenyl.


In one embodiment, R4a is a 5-6 membered heteroaryl ring having 1-3 ring heteroatoms independently selected from N, S and O and optionally substituted with 1-2 substituents independently selected from (1-6C)alkyl, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2— (3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, and (1-3C trifluoroalkoxy)(1-3C)trifluoroalkyl. In one embodiment, R4a is pyridyl, pyrimidinyl pyridazinyl, pyrazolyl, imidazolyl, thionyl, 1,2,4-triazolyl, 1,2,3-triazolyl, thiazolyl, oxazolyl, 1,3,4-oxadiazolyl, 1,2,4-oxadiazolyl or imidazo[1,2-a]pyridinyl optionally substituted with 1-2 substituents independently selected from (1-6C)alkyl, hydroxy(1-6C)alkyl, trifluoro(1-6C)alkyl, (3-6C)cycloalkyl, (3-6C cycloalkyl)CH2— (3-6C cycloalkyl)C(═O)—, (1-3C alkoxy)(1-6C)alkyl, (1-6C)alkoxy, (1-6C)alkylsulfonyl, NH2, (1-6C alkyl)amino, di(1-6C alkyl)amino, and (1-3C trifluoroalkoxy)(1-3C)trifluoroalkyl. In one embodiment, R4a is pyrazinyl.


In one embodiment, R5a is as defined for Formula I.


In one embodiment, R5a is selected from hydrogen, halogen, (1-6C)alkyl and phenyl.


In one embodiment, R5a is hydrogen.


In one embodiment, R5a is halogen.


In one embodiment, R5a is (1-6C)alkyl. In one embodiment, R5a is methyl.


In one embodiment, R5a is phenyl.


In one embodiment, Ring C is formula C-2, in which R3a is (1-6C)alkyl, trifluoro(1-6C)alkyl or phenyl; R4a is (1-6C)alkyl, trifluoro(1-6C)alkyl, phenyl or pyrazinyl; and R5a is hydrogen, (1-6C)alkyl or phenyl.


In one embodiment, Formula I includes compounds of Formula Ia, wherein D is S or O; X is O; Ring C is C-1; R3 is Ar2; R4 is hetAr4; R5 is (1-6C)alkyl; and R1, R2, Ar2 and hetAr4 are as defined for Formula I. In one embodiment of Formula I-a, R3 is phenyl.


In one embodiment of Formula Ia, D is S or O; X is O; Ring C is C-1; R3 is Ar2; R4 is pyrazolyl optionally substituted with one or more groups independently selected from (1-6C)alkyl; R5 is (1-6C)alkyl; and R1, R2, Ar2 is as defined for Formula I. In one embodiment of Formula I-a, R3 is phenyl.


It will be appreciated that certain compounds according to the invention may contain one or more centers of asymmetry and may therefore be prepared and isolated in a mixture of isomers such as a racemic mixture, or in an enantiomerically pure form.


It will further be appreciated that the compounds of Formula I or their salts may be isolated in the form of solvates, and accordingly that any such solvate is included within the scope of the present invention. For example, compounds of Formula I can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.


The compounds of Formula I include pharmaceutically acceptable salts thereof. In addition, the compounds of Formula I also include other salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which are useful as intermediates for preparing and/or purifying compounds of Formula I and/or for separating enantiomers of compounds of Formula I. Particular examples of salts include hydrochloride salts and trifluoroacetate salts.


In one embodiment, the compounds of Formula I include the free base form of compounds of Examples 1-5, or a pharmaceutically acceptable salt thereof.


In one embodiment, the compounds of Formula I include the hydrochloride salts of compounds of Examples 1-5.


In one embodiment, the compounds of Formula I include the trifluoroacetate salts of compounds of Examples 1-5.


The term “pharmaceutically acceptable” indicates that the substance or composition is compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.


The present invention also provides a process for the preparation of a compound of Formula I or a salt thereof as defined herein, which comprises:


(a) for a compound of Formula I where X is O, coupling a corresponding compound having the formula II




embedded image


with a corresponding compound having the formula III




embedded image


in the presence carbonyldiimidazole or triphosgene and a base; or


(b) for a compound of Formula I where X is S, coupling a corresponding compound having the formula II




embedded image


with a corresponding compound of formula III




embedded image


in the presence di(1H-imidazol-2-yl)methanethione and a base; or


(c) for a compound of Formula I where X is O, coupling a corresponding compound having the formula II




embedded image


with a corresponding compound having the formula IV




embedded image


where L1 is a leaving group, in the presence of a base; or


(d) for a compound of Formula I where X is O, coupling a corresponding compound having the formula V




embedded image


where L2 is a leaving group, with a corresponding compound having the formula III




embedded image


in the presence of a base; or


(e) for a compound of Formula I where X is O, activating a corresponding compound having the formula VI




embedded image


with diphenylphosphoryl azide followed by coupling the activated intermediate with a corresponding compound having the formula III




embedded image


in the presence a base; or


(f) for a compound of Formula I where X is O, coupling a corresponding compound having the formula II




embedded image


with a corresponding compound having the formula VII




embedded image


in the presence of a base; or


(g) for a compound of Formula I where X is O, coupling a corresponding compound having the formula VIII




embedded image


with a corresponding compound having the formula III




embedded image


in the presence of a base; and


optionally removing protecting groups and optionally preparing a pharmaceutically acceptable salt thereof.


In the above methods, the term “corresponding” means that the definitions for the “corresponding compound” are as defined for Formula I unless stated otherwise.


Referring to method (a), the base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include dichloromethane, dichloroethane, THF, DMA and DMF. The reaction is conveniently performed at ambient temperature.


Referring to method (b), the base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include dichloromethane, dichloroethane, THF, DMA and DMF. The reaction is conveniently performed at ambient temperature.


Referring to method (c), the leaving group may be, for example, phenoxy or 4-nitrophenoxy. The base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include DMA, DMF and DCE. The reaction is conveniently performed at ambient temperature or elevated temperature, for example at 60° C.


Referring to method (d), the leaving group may be, for example, phenoxy or 4-nitrophenoxy. The base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include DCE, DMA and DMF. The reaction is conveniently performed at ambient temperature.


Referring to method (e), the base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include toluene and DMF. The reaction is conveniently performed at elevated temperatures, for example the reflux temperature of the solvent.


Referring to methods (f) and (g), the base may be an amine base, such as triethylamine or diisopropylethylamine. Suitable solvents include DCM, DCE, DMF and THF. The reaction is conveniently performed at temperatures between about 0° C. and ambient temperature.


Amine groups in compounds described in any of the above methods may be protected with any convenient amine protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2nd ed. New York; John Wiley & Sons, Inc., 1991. Examples of amine protecting groups include acyl and alkoxycarbonyl groups, such as t-butoxycarbonyl (BOC) and [2-(trimethylsilyl)ethoxy]methyl (SEM). Likewise, carboxyl groups may be protected with any convenient carboxyl protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2nd ed. New York; John Wiley & Sons, Inc., 1991. Examples of carboxyl protecting groups include (1-6C)alkyl groups, such as methyl, ethyl and t-butyl. Alcohol groups may be protected with any convenient alcohol protecting group, for example as described in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2nd ed. New York; John Wiley & Sons, Inc., 1991. Examples of alcohol protecting groups include benzyl, trityl, silyl ethers, and the like.


The compounds of the formulas II, V, VI, VII, and VIII are also provided as further aspects of the invention. In one embodiment, compounds of the formulas II, V, VI, VII, and VIII are useful as intermediates for the preparation of compounds of Formula I.


Compounds of Formula I are useful in the treatment of pain, cancer, inflammation/inflammatory diseases, neurodegenerative diseases, certain infectious diseases, Sjogren's syndrome, endometriosis, diabetic peripheral neuropathy, prostatitis or pelvic pain syndrome.


In one embodiment, compounds of Formula I are useful for treating pain, including chronic and acute pain. For example, compounds of Formula I are useful in the treatment of multiple types of pain including inflammatory pain, neuropathic pain, and pain associated with cancer, surgery or bone fracture.


In one embodiment, compounds of Formula I are useful for treating acute pain. Acute pain, as defined by the International Association for the Study of Pain, results from disease, inflammation, or injury to tissues. This type of pain generally comes on suddenly, for example, after trauma or surgery, and may be accompanied by anxiety or stress, and is confined to a given period of time and severity. In some instances, it can become chronic.


In one embodiment, compounds of Formula I are useful for treating chronic pain. Chronic pain, as defined by the International Association for the Study of Pain, is widely believed to represent a disease in itself. It can be made much worse by environmental and psychological factors. Chronic pain persists over a longer period than acute pain and is resistant to most medical treatments, generally over 3 months or more. It can and often does cause severe problems for patients.


Compounds of Formula I are also useful for treating cancer. Particular examples include neuroblastoma, ovarian, pancreatic, colorectal and prostate cancer.


Compounds of Formula I are also useful for treating inflammation and certain infectious diseases. For example, compounds of Formula I may be used to treat interstitial cystitis (IC), painful bladder syndrome (PBS), urinary incontinence, asthma, atopic dermatitis, and psoriasis.


Compounds of Formula I are also useful for treating a neurodegenerative disease in a mammal, comprising administering to said mammal one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said neurodegenerative disease. In one embodiment, compounds of Formula I may also be used to treat demyelination and dysmyelination by promoting myelination, neuronal survival, and oligodendrocyte differentiation via blocking Sp35-TrkA interaction. In one embodiment, the neurodegenerative disease is multiple sclerosis. In one embodiment, the neurodegenerative disease is Parkinson's disease. In one embodiment, the neurodegenerative disease is Alzheimer's disease.


Compounds of Formula I are also useful for treating certain infectious diseases such as Trypanosoma cruzi infection in a mammal.


Compounds of Formula I are also useful for treating Sjogren's syndrome in a mammal.


Compounds of Formula I are also useful for treating endometriosis in a mammal.


Compounds of Formula I are also useful for treating diabetic peripheral neuropathy in a mammal.


Compounds of Formula I are also useful for treating prostatitis in a mammal.


Compounds of Formula I are also useful for treating pelvic pain syndrome in a mammal.


Compounds of Formula I are also useful in treating diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis, and bone metastases.


As used herein, terms “treat” or “treatment” refer to therapeutic or palliative measures. Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disorder or condition, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.


In certain embodiments, compounds of Formula I are useful for preventing diseases and disorders as defined herein. The term “preventing” as used herein means the prevention of the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein, or a symptom thereof, and includes to the administration of a compound of Formula I prior to the onset of symptoms.


Accordingly, one embodiment of this invention provides a method of treating pain in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said pain. In one embodiment, the pain is chronic pain. In one embodiment, the pain is acute pain. In one embodiment, the pain is inflammatory pain, neuropathic pain, or pain associated with cancer, surgery, or bone fracture.


Another embodiment of this invention provides a method of preventing pain in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to prevent said pain. In one embodiment, the pain is chronic pain. In one embodiment, the pain is acute pain. In one embodiment, the pain is inflammatory pain, neuropathic pain, or pain associated with cancer, surgery, or bone fracture.


Another embodiment of this invention provides a method of treating cancer in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said cancer.


In one embodiment, provided herein is a method for treating a patient diagnosed with a cancer having a dysregulation of TrkA, comprising administering to the patient a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof.


In one embodiment, the dysregulation of TrkA comprises overexpression of wild-type TrkA (autocrine activation).


In one embodiment, the dysregulation of TrkA comprises one or more chromosome translocations or inversions resulting in TrkA gene fusions. In one embodiment, the dysregulation is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from non-TrkA and TrkA proteins, and at a minimum the TrkA kinase domain. In one embodiment, the TrkA fusion protein is LMNA-TrkA, TFG-TrkA, TPM3-TrkA, CD74-TrkA, NFASC-TrkA, MPRIP-TrkA, BCAN-TrkA, or TPR-TrkA, where:


LMNA=Prelamin-A/C;


TFG=TRK-fused gene protein;


TPM3=Tropomysin alpha-3;


CD74=HLA class II histocompatibility antigen gamma chain;


NFASC=Neurofascin;


MPRIP=MPRIP protein;


BCAN=Brevican core protein; and


TPR=Nucleoprotein TPR


In one embodiment, the dysregulation of TrkA comprises one or more deletions, insertions or mutations in the TrkA protein. In one embodiment, the dysregulation comprises a deletion of one or more residues from the TrkA protein, resulting in constitutive activity of TrkA kinase. In one embodiment the deletion includes deletion of residues 303-377 in TrkA Isoform 2.


In one embodiment, the dysregulation of TrkA comprises a splice variation in which the expressed protein is an alternatively spliced variant of TrkA having one or more residues deleted resulting in constitutive activity of TrkA kinase. In one embodiment, an alternatively spliced form of TrkA with constitutive activity has deletions of exons 8, 9, and 11 resulting in an expressed protein missing residues 192-284 and 393-398 relative to TrkA Isoform 2.


Cancers identified as having dysregulation of TrkA (see literature references below; also see www.cancer.gov and www.nccn.org) include:


(A) Cancers wherein the dysregulation of TrkA comprises one or more chromosome translocations or inversions resulting in TrkA gene fusions, including:














Cancer
Literature reference(s)
Standard of Care







Non-Small Cell
Vaishnavi et al. 2013:
radiotherapy (e.g. radioiodide therapy,


Lung Cancer
Nature Medicine 19,
external-beam radiation, radium 223



1469-1472
therapy), chemotherapeutics as single




agents (e.g. afatinib dimaleate,




bevacizumab, carboplatin, cetuximab,




cisplatin, crizotinib, erlotinib, gefitinib,




gemcitabine, methotrexate, paclitaxel,




pemetrexed) or combinations (e.g.




carboplatin-paclitaxel, gemcitabine-




paclitaxel, chemoradiation)


Papillary Thyroid
Caria et al. 2010: Cancer
Radiotherapies (e.g. radioiodide therapy,


Carcinoma
Genetics and
external-beam radiation) and



Cytogenetics 203: 21-29
chemotherapeutics (e.g. sorafenib,




sunitinib, pazopanib)


Glioblastoma
Frattini et al. 2013:
Chemotherapeutics (e.g. bevacizumab,


Multiforme
Nature Genet.
everolimus, lomustine, temozolomide)



45(10): 1141-9


Colorectal
Martin-Zanca et al.
Chemotherapeutics as single agents


Carcinoma
1986: Nature 319: 743
(aflibercept, bevacizumab, capecitabine,




cetuximab, fluorouracil, irinotecan,




leucovorin, oxaliplatin, panitumumab,




regorafenib) or combinations (e.g. folfox,




folfiri, capox, folfiri-bevacizumab, folfiri-




cetuximab, xelox)


Melanoma
WO 2013/059740 A1
Chemotherapeutics (e.g. aldesleukin,




dabrafenib, dacarbazine, interferon alfa-




2b, ipilimumab, peginterferon alfa-2b,




trametinib, vemurafenib)









(B) Cancers wherein the dysregulation of TrkA comprises one or more deletions, insertions or mutations in the TrkA protein, including:














Cancer
Literature reference(s)
Standard of care







Acute Myeloid
Meyer 2007: Leukemia 21:
Chemotherapeutics as single


leukemia
2171-2180
agents (e.g. arsenic trioxide,



Reuther et al. 2000: Mol Cell
cyclophosphamide, cytarabine,



Biol 20: 8655-8666
daunorubicin, doxorubicin,




vincristine) or combinations (e.g.




ADE)


Large Cell
Marchetti et al 2008: Human
Radiotherapy (e.g. radioiodide


Neuroendocrine
Mutation 29(5): 609-616
therapy, external-beam radiation,


Carcinoma

radium 223 therapy) and/or




chemotherapeutics (e.g. cisplatin,




carboplatin, etoposide)


Neuroblastoma
Tacconelli et al 2004: Cancer
Chemotherapeutics (e.g.



Cell 6: 347
cyclophosphamide, doxorubicin,




vincristine)









(C) Cancers driven by overexpression of wild-type TrkA (autocrine activation), including:














Cancer
Literature Reference(s)
Standard of care







Prostate Carcinoma
Walch et al: Clinical &
Radiotherapy (e.g. radium 223



Experimental Metastasis 17: 307-314
therapy) or chemotherapeutics



Papatsoris et al 2007: Expert
(e.g. abiraterone, cabazitaxel,



Opinion on Investigational Drugs
degarelix, denosumab, docetaxel,



16(3): 303-309
enzalutamide, leuprolide,




prednisone, sipuleucel-T)


Neuroblastoma
Van Noesel et al 2004: Gene 325:
Chemotherapeutics (e.g.



1-15
cyclophosphamide, doxorubicin,




vincristine)


Pancreatic
Zhang et al 2005: Oncology
Chemotherapeutics as single


Carcinoma
Reports 14: 161-171
agents (e.g. erlotinib,




fluorouracil, gemcitabine,




mitomycin C) or combinations




(e.g. gemcitabine-oxaliplatin)


Melanoma
Truzzi et al 2008: Journal of
Chemotherapeutics (e.g.



Investigative Dermatology
aldesleukin, dabrafenib,



128(8): 2031
dacarbazine, interferon alfa-2b,




ipilimumab, peginterferon alfa-




2b, trametinib, vemurafenib)


Head and Neck
Kolokythas et al 2010: Journal of
Radiotherapy and/or


Squamous Cell
Oral and Maxillofacial Surgery
chemotherapeutics (e.g.


Carcinoma
68(6): 1290-1295
bleomycin, cetuximab, cisplatin,




docetaxel, fluorouracil,




methotrexate)


Gastric Carcinoma
Ni et al 2012: Asian Pacific
Chemotherapeutics (e.g.



Journal of Cancer Prevention 13:
docetaxel, doxorubucin,



1511
fluorouracil, mitomycin C,




trastuzumab)









In one embodiment, provided herein is a method for treating a patient diagnosed with a cancer having a dysregulation of TrkA, comprising administering to the patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, wherein the cancer is selected from non-small cell lung cancer, papillary thyroid carcinoma, glioblastoma multiforme, acute myeloid leukemia, colorectal carcinoma, large cell neuroendocrine carcinoma, prostate cancer, neuroblastoma, pancreatic carcinoma, melanoma, head and neck squamous cell carcinoma and gastric carcinoma.


In one embodiment, the compounds of the present invention are useful for treating cancer in combination with one or more additional therapeutic agents or therapies that work by the same or a different mechanism of action.


In one embodiment, the additional therapeutic agent(s) is selected from receptor tyrosine kinase-targeted therapeutic agents, including cabozantinib, crizotinib, erlotinib, gefitinib, imatinib, lapatinib, nilotinib, pazopanib, pertuzumab, regorafenib, sunitinib, and trastuzumab.


In one embodiment, the additional therapeutic agent(s) is selected from signal transduction pathway inhibitors, including Ras-Raf-MEK-ERK pathway inhibitors (e.g. sorafenib, trametinib, vemurafenib), PI3K-Akt-mTOR-S6K pathway inhibitors (e.g. everolimus, rapamycin, perifosine, temsirolimus) and modulators of the apoptosis pathway (e.g. obataclax).


In one embodiment, the additional therapeutic agent(s) is selected from cytotoxic chemotherapeutics, including arsenic trioxide, bleomycin, cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, cytarabine, dacarbazine, daunorubicin, docetaxel, doxorubicin, etoposide, fluorouracil, gemcitabine, irinotecan, lomustine, methotrexate, mitomycin C, oxaliplatin, paclitaxel, pemetrexed, temozolomide, and vincristine.


In one embodiment, the additional therapeutic agent(s) is selected from angiogenesis-targeted therapies, including aflibercept and bevacizumab.


In one embodiment, the additional therapeutic agent(s) is selected from immune-targeted agents, including aldesleukin, ipilimumab, lambrolizumab, nivolumab, sipuleucel-T.


In one embodiment, the additional therapeutic agent(s) is selected from agents active against the TrkA pathway, including NGF-targeted biopharmaceuticals such as NGF antibodies, and panTrk inhibitors.


In one embodiment, the additional therapeutic agent or therapy is radiotherapy, including radioiodide therapy, external-beam radiation and radium 223 therapy.


In one embodiment, the additional therapeutic agent(s) includes any one of the above listed therapies or therapeutic agents which are standards of care in cancers wherein the cancer has a dysregulation of TrkA.


In one embodiment, provided herein is a method of treating cancer in a patient, comprising administering to said patient a compound of the invention or a pharmaceutically acceptable salt thereof, in combination with at least one additional therapy or therapeutic agent selected from radiotherapy (e.g. radioiodide therapy, external-beam radiation, radium 223 therapy), cytotoxic chemotherapeutics (e.g. arsenic trioxide, bleomycin, cabazitaxel, capecitabine, carboplatin, cisplatin, cyclophosphamide, cytarabine, dacarbazine, daunorubicin, docetaxel, doxorubicin, etoposide, fluorouracil, gemcitabine, irinotecan, lomustine, methotrexate, mitomycin C, oxaliplatin, paclitaxel, pemetrexed, temozolomide, vincristine), tyrosine kinase targeted-therapeutics (e.g. afatinib, cabozantinib, cetuximab, crizotinib, dabrafenib, erlotinib, gefitinib, imatinib, lapatinib, nilotinib, pazopanib, panitumumab, pertuzumab, regorafenib, sunitinib, trastuzumab), apoptosis modulators and signal transduction inhibitors (e.g. everolimus, perifosine, rapamycin, sorafenib, temsirolimus, trametinib, vemurafenib), immune-targeted therapies (e.g. aldesleukin, interferon alfa-2b, ipilimumab, lambrolizumab, nivolumab, prednisone, sipuleucel-T) and angiogenesis-targeted therapies (e.g. aflibercept, bevacizumab), wherein the amount of the compound of the invention or a pharmaceutically acceptable salt thereof is, in combination with the additional therapy or therapeutic agent, is effective in treating said cancer. These additional therapeutic agents may be administered with one or more compounds of the invention as part of the same or separate dosage forms, via the same or different routes of administration, and on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.


Also provided herein is (i) a pharmaceutical combination for treating cancer in a patient in need thereof, which comprises (a) a compound of the invention or a pharmaceutically acceptable salt thereof, (b) an additional therapeutic agent and (c) optionally at least one pharmaceutically acceptable carrier for simultaneous, separate or sequential use for the treatment of a tumor disease, wherein the amounts of the compound or salt thereof and of the additional therapeutic agent are together effective in treating said cancer; (ii) a pharmaceutical composition comprising such a combination; (iii) the use of such a combination for the preparation of a medicament for the treatment of cancer; and (iv) a commercial package or product comprising such a combination as a combined preparation for simultaneous, separate or sequential use; and to a method of treatment of cancer a patient in need thereof.


In one embodiment, the combination therapy is for treating a cancer is selected from non-small cell lung cancer, papillary thyroid carcinoma, glioblastoma multiforme, acute myeloid leukemia, colorectal carcinoma, large cell neuroendocrine carcinoma, prostate cancer, neuroblastoma, pancreatic carcinoma, melanoma, head and neck squamous cell carcinoma and gastric carcinoma.


Another embodiment of this invention provides a method of treating inflammation or an inflammatory disease or disorder in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said inflammation. In one embodiment, the inflammatory disease is inflammatory lung diseases (such as asthma), interstitial cystitis, bladder pain syndrome, inflammatory bowel diseases (including ulcerative colitis and Crohn's disease), and inflammatory skin diseases such as atopic dermatitis.


In one embodiment, the method of treating inflammation or an inflammatory disease or disorder comprises administering a compound of the invention in combination with one or more additional agents. Examples of additional agents include anti-TNF treatments (for example monoclonal antibody such as infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), and golimumab (Simponi), or a circulating receptor fusion protein such as etanercept (Enbrel)), antimetabolite and antifolate drug (for example Methotrexate), or targeted kinase inhibitors (for example JAK family inhibitors Ruxolitinib, Tofacitinib, CYT387, Lestaurtinib, Pacritinib and TG101348).


Another embodiment of this invention provides a method of treating Trypanosoma cruzi infection in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said Trypanosoma cruzi infection.


Another embodiment of this invention provides a method of treating Sjogren's syndrome in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said syndrome.


Another embodiment of this invention provides a method of treating endometriosis in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said endometriosis.


Another embodiment of this invention provides a method of treating diabetic peripheral neuropathy in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said diabetic peripheral neuropathy.


Another embodiment of this invention provides a method of treating prostatitis in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said prostatitis.


Another embodiment of this invention provides a method of treating pelvic pain syndrome in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said pelvic pain syndrome.


Another embodiment of this invention provides a method of treating a neurodegenerative disease in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said neurodegenerative disease.


As used herein, an “effective amount” means an amount of compound that, when administered to a mammal in need of such treatment, is sufficient to (i) treat a particular disease, condition, or disorder which can be treated with a compound of Formula I, or (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder described herein.


The amount of a compound of Formula I that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.


As used herein, the term “mammal” refers to a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, and primates, including humans.


The compounds of the present invention can be used in combination with one or more additional therapeutic agents that work by the same or a different mechanism of action. Examples of additional therapeutic agents include anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), and opioids (such as morphine), and chemotherapeutic agents.


Also provided herein is a pharmaceutical combination comprising an effective amount of: (a) at least one compound of Formula I; and (b) at least one additional therapeutic agent selected from anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), and opioids (such as morphine), for use in the treatment of pain in a mammal, wherein (a) and (b) can be in separate dosage forms or in the same dosage form.


The term “pharmaceutical combination” as used herein refers to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that at least one of the compounds of Formula I, and at least one additional therapeutic agent are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that at least one of the compounds of Formula I, and at least one additional therapeutic agent, are administered to a patient as separate entities either simultaneously or sequentially with variable intervening time limits, wherein such administration provides effective levels of the two or more compounds in the body of the patient. These also apply to cocktail therapies, e.g. the administration of three or more active ingredients.


Also provided herein is a method of treating pain in a mammal, comprising co-administering to a mammal in need thereof an effective amount of: (a) at least one compound of Formula I; and (b) at least one additional therapeutic agent selected from anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), opioids (such as morphine), calcitonin gene-related peptide receptor antagonists, subtype-selective ion channel modulators, anticonvulsants (for example Pregabalin and gabapentin), dual serotonin-norepinephrin reuptake inhibitors (for example duloxetine, venlafaxine and milnacipran), and tricyclic antidepressants (such as amitriptyline, nortriptyline and desipramine).


Another embodiment of this invention provides a method of treating diseases related to an imbalance of the regulation of bone remodeling in a mammal, comprising administering to said mammal in need thereof one or more compounds of Formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat said disease. In one embodiment, the disease is osteoporosis, rheumatoid arthritis, and bone metastases.


In one embodiment, the method for treating diseases related to an imbalance of the regulation of bone remodeling in a mammal comprises administering a TrkA inhibitor of the invention in combination with one or more additional therapeutic agents or therapies. Examples of additional therapeutic agents or therapies include anti-TNF treatments (for example monoclonal antibody such as infliximab (Remicade), adalimumab (Humira), certolizumab pegol (Cimzia), and golimumab (Simponi), or with a circulating receptor fusion protein such as etanercept (Enbrel)), antimetabolite and antifolate drug (for example Methotrexate), or targeted kinase inhibitors (for example JAK family inhibitors Ruxolitinib, Tofacitinib, CYT387, Lestaurtinib, Pacritinib and TG101348).


The term “co-administering” is meant to encompass administration of the selected therapeutic agents to a single patient, and is intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different times. This term encompasses administration of two or more agents to a mammal so that both agents and/or their metabolites are present in the mammal at the same time. It includes simultaneous administration in separate compositions, administration at different times in separate compositions, and/or administration in a composition in which both agents are present. In some embodiments, the compound(s) of the invention and the other therapeutic agent(s) are administered in a single composition. In some embodiments, compound(s) of the invention and the other agent(s) are admixed in the composition.


Also provided herein is a medicament containing a compound of Formula I for treatment of pain in a mammal in combination with an additional therapeutic agent selected from anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), and opioids (such as morphine).


Also provided herein is a medicament containing a therapeutic agent selected from anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), and opioids (such as morphine) for treatment of pain in a mammal in combination with a compound of Formula I.


Compounds of the invention may be administered by any convenient route, e.g. into the gastrointestinal tract (e.g. rectally or orally), the nose, lungs, musculature or vasculature, or transdermally or dermally. Compounds may be administered in any convenient administrative form, e.g. tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g. diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. If parenteral administration is desired, the compositions will be sterile and in a solution or suspension form suitable for injection or infusion. Such compositions form a further aspect of the invention.


Another formulation may be prepared by mixing a compound described herein and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound described herein or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).


Accordingly, another aspect of the present invention provides a pharmaceutical composition, which comprises a compound of Formula I or a pharmaceutically acceptable salt thereof, as defined hereinabove, together with a pharmaceutically acceptable diluent or carrier.


According to another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of pain in a mammal. In one embodiment, the pain is chronic pain. In one embodiment the pain is acute pain. In one embodiment, the pain is inflammatory pain, neuropathic pain, or pain associated with cancer, surgery, or bone fracture.


According to another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer in a mammal.


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of inflammation or an inflammatory disease or disorder in a mammal. In one embodiment, the inflammatory disease is inflammatory lung diseases (such as asthma), interstitial cystitis, bladder pain syndrome, inflammatory bowel diseases (including ulcerative colitis and Crohn's disease), and inflammatory skin diseases such as atopic dermatitis.


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of infectious diseases, for example Trypanosoma cruzi infection, in a mammal.


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of Sjogren's syndrome in a mammal.


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of endometriosis in a mammal.


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of diabetic peripheral neuropathy in a mammal,


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of prostatitis in a mammal,


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of pelvic pain syndrome in a mammal,


In another embodiment, the present invention provides a compound of Formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of a neurodegenerative disease in a mammal.


According to a further aspect, the present invention provides the use of a compound of Formula I or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a condition selected from pain, cancer, inflammation, neurodegenerative disease or Trypanosoma cruzi infection. In one embodiment, the condition is chronic pain. In one embodiment, the condition is acute pain. In one embodiment, the pain is inflammatory pain, neuropathic pain, or pain associated with cancer, surgery, or bone fracture. In one embodiment, the condition is cancer. In one embodiment, the condition is inflammation. In one embodiment, the condition is a neurodegenerative disease. In one embodiment, the condition is Trypanosoma cruzi infection. In one embodiment, the condition is Sjogren's syndrome. In one embodiment, the condition is endometriosis. In one embodiment, the condition is diabetic peripheral neuropathy. In one embodiment, the condition is prostatitis. In one embodiment, the condition is pelvic pain syndrome.


EXAMPLES

The following examples illustrate the invention. In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Lancaster, TCI or Maybridge, and were used without further purification unless otherwise indicated.


The reactions set forth below were done generally under a positive pressure of nitrogen or argon (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.


Column chromatography was done on a Biotage system (Manufacturer: Dyax Corporation) having a silica gel or C-18 reverse phase column, or on a silica SepPak cartridge (Waters).


Biological Assays
Example A-1
TrkA Kinase Binding Assay

TrkA binding activity was determined in a TrkA LanthaScreen™ Eu Kinase Binding Assay. 5 nM His-tagged recombinant human TrkA (6HIS tagged cytoplasmic domain from Invitrogen, Cat. No. PV3144) was incubated with 4 nM Alexa-Fluor® Tracer 236 (Invitrogen Cat. No. PV5592), 2 nM biotinylated anti-His (Invitrogen Cat. No. PV6090), and 2 nM europium-labeled Streptavidin (Invitrogen Cat. No. PV5899), in buffer (25 mM MOPS, pH 7.5, 5 mM MgCl2, 0.005% Triton X-100). Three fold serial dilutions of compounds of the invention in DMSO were added to a final percentage of 2% DMSO. After 60-minute incubation at 22° C., the reaction was measured using the EnVision mutlimode plate reader (PerkinElmer) via TR-FRET dual wavelength detection at 615 nM and 665 nM. The percent of control was calculated using a ratiometric emission factor. The IC50 values were determined by fitting a four parameter model to the percent of control data.


Table A provides averaged IC50 values for compounds of the invention when tested in the assay of Example A, where A represents an averaged IC50 value <100 nM; and B represents an averaged IC50 value from 100 to 1,000 nM.












TABLE A








TrkA enzyme IC50



Ex. #
(nM)









1
A



2
A



3
B



4
B



5
A










Example A-2
p38 Kinase Binding Assay

p38α binding activity was determined in a p38α LanthaScreen™ Eu Kinase Binding Assay. 5 nM of inactive, GST-tagged recombinant human p38α (GST-tagged cytoplasmic domain from Invitrogen, Catalog No. PV3305) was incubated with 5 nM Alexa-Fluor® Tracer 199 (Invitrogen Cat. No. PV5830), and 2 nM europium labeled anti-GST antibody (Invitrogen Cat. No. PV5594), in buffer (25 mM [Na+] HEPES pH 7.3, 10 mM MgCl2, 100 μM NaVO4). Three fold serial dilutions of compounds of the invention in DMSO were added to a final percentage of 2% DMSO. After 60-minute incubation at 22° C., the reaction was measured using the EnVision multimode plate reader (PerkinElmer) via TR-FRET dual wavelength detection at 615 nM and 665 nM. The percent of control was calculated using a ratiometric emission factor. The IC50 values were determined by fitting a four parameter model to the percent of control data. The compounds of Examples 1-5 were tested in this assay, and all compounds were found to be 1000 fold more potent against TrkA than p38α.


Example B
Off-Target Kinase Profiling

A representative compound of the invention (Example 2) was tested for off-target kinase activity at a concentration of 10 μM by Millipore, Inc. in their KinaseProfiler™ service against all the kinases available in their full kinase panel. The compound was run in duplicate at a concentration of ATP near the Km for each individual kinase according to Millipore's specifications. The results are shown in Table B. Data are reported as percent of control (POC) and are the average of the two replicates.


In the KinaseProfiler™ the representative compound showed remarkable and unexpected selectivity for inhibiting TrkA and TrkB versus other kinases in the panel. In fact, the compound was largely inactive against off-target kinases at a concentration of 10 μM, and thus would not be expected to inhibit off-target kinases at therapeutic doses in mammals. The ability of compounds of the invention to selectively inhibit the Trk pathway without inhibiting other off-target kinases could translate into drug profiles that are essentially free of side-effects related to inhibition of off-target kinases. Such a drug profile would represent a safer approach to treating pain, inflammation, cancer and certain skin diseases than has been previously reported.












TABLE B








Example 2



Kinase
Avg POC



















Abl2
136



Abl-P
148



AKT1
133



AKT2
171.5



AKT3
116



ALK
108



ALK4
107



AMPK(A1/B1/G1)
134.5



ARK5
102



AURKA
116



Axl
122.5



BLK_m
112



Bmx
129.5



BrSK1
132.5



BrSK2
116.5



BTK
123



CAMK1
95.5



CAMK1d
132.5



CAMK2b
102



CAMK2d
118



CAMK2g
103



CAMK4
132.5



CDK1/cyclinB
106



CDK2/cyclinA
122



CDK2/cyclinE
100



CDK3/cyclinE
105



CDK5/p25
104.5



CDK5/p35
111



CDK6/cyclinD3
119.5



CDK7/cyclinH/MAT1
113



CDK9/cyclinT1
108



CHK1
126.5



CHK2
133



CK1_y
103



CK1delta
117.5



CK1gamma1
104



CK1gamma2
110



CK1gamma3
114



CK2
95



CK2alpha2
104



CLK2
118



CLK3
110.5



c-RAF
96.5



CSK
136



DAPK1
126.5



DAPK2
103.5



DAPK3
120



DCAMKL2
210.5



DDR2
98



DMPK
107.5



DRAK1
119



DYRK2
88



eEF-2K
151.5



EGFR
111



EphA1
81.5



EphA2
127



EphA3
120



EphA4
109



EphA5
118.5



EphA7
103



EphA8
118.5



EphB1
136



EphB2
119



EphB3
72.5



EphB4
130



ErbB4
125



ERK1
96.5



ERK2
135



FAK
106



FAK2
107



Fer
78.5



Fes
121.5



FGFR1
80.5



FGFR2
116.5



FGFR3
117



FGFR4
141.5



Fgr
128



Flt1
104



Flt3
98



Flt4
107



Fms
89.5



Fyn
105.5



GRK5
82.5



GRK6
98.5



GRK7
112



GSK3alpha
147.5



GSK3beta
127.5



Haspin
112.5



Hck
134.5



HIPK1
108



HIPK2
98.5



HIPK3
105.5



IGF-1R
82.5



IGF-1R Activated
97.5



IKKalpha
133



IKKbeta
112



IR
99



IR Activated
113.5



IRAK1
107.5



IRAK4
112



IRR
109



ITK
125.5



JAK2
147.5



JAK3
113



JNK1alpha1
113



JNK2alpha2
97.5



JNK3
120



KDR
126



KIT
102.5



Lck
103



LIMK1
102



LKB1
96.5



LOK
110.5



Lyn
103.5



MAP3K5
107.5



MAP4K2
121.5



MAPKAP-K2
143.5



MAPKAP-K3
116



MAPKAP-K5
95.5



MARK1
107



MARK2
100.5



MEK1
103.5



MELK
99



Mer
105.5



Met
120



MINK
117.5



MKK4_m
73.5



MKK6
134



MKK7beta
143.5



MKNK2
95



MLK1
95.5



MRCKalpha
147.5



MRCKbeta
113



MSK1
151



MSK2
136



MSSK1
132



MST1
100.5



MST2
109



MST3
157.5



mTOR
92.5



mTOR/FKBP12
103



MuSK
88



MYLK
99



NEK11
73.5



NEK2
122.5



NEK3
108.5



NEK6
117.5



NEK7
119.5



NLK
125.5



p38alpha
91



p38beta
106.5



p38delta
104.5



p38gamma
105



p70S6K
322



PAK2
98.5



PAK4
103.5



PAK5
131.5



PAK6
135



PASK
113



PDGFRalpha
102.5



PDGFRbeta
162



PDK1
115



PhKgamma2
108



Pim-1
109.5



Pim-2
142.5



Pim-3
107.5



PKAC-alpha
134



PKCalpha
102.5



PKCbetaI
102



PKCbetaII
95



PKCdelta
97



PKCepsilon
102



PKCeta
103.5



PKCgamma
90



PKCiota
67



PKCtheta
130



PKCzeta
108



PKD1
97.5



PKD2
110.5



Plk1
103.5



Plk2
99



Plk3
105



PRK2
104.5



PRKG1alpha
118



PRKG1beta
119



PrKX
122



PTK5
104.5



PTK6
113.5



Ret
91.5



RIPK2
98



ROCK-I
113



ROCK-II
115.5



Ron
110.5



Ros
96.5



Rse
108



Rsk1
123.5



Rsk2
131



Rsk3
99



Rsk4
98



SGK1
139.5



SGK2
144.5



SGK3
151



SIK
145



SRC
110.5



SRPK1
114



SRPK2
113.5



STK33
120.5



Syk
127.5



TAK1
95.5



TAO1
103.5



TAO2
123.5



TAO3
94



TBK1
100.5



TEC Activated
131



Tie2
131.5



TLK2
98



TNK2
142.5



TrkA
−1



TrkB
3



TSSK1
92



TSSK2
142.5



Txk
163



ULK2
91



ULK3
90



VRK2
91.5



WNK2
115



WNK3
112.5



Yes
128.5



ZAP-70
133










Preparation of Synthetic Intermediates
Intermediate 1



embedded image


1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine
Step A: ethyl 1-methyl-1H-pyrazole-4-carboxylate

To a 3000-mL three-necked flask was added ethyl 2-formyl-3-oxopropanoate (100 g, 694 mmol), followed by anhydrous 200-proof EtOH (694 mL). The reaction was cooled in an ice bath to 5° C., and then methylhydrazine (35.8 mL, 680 mmol) was added dropwise. A vigorous exotherm was observed during hydrazine addition and the temperature was kept below 12° C. by controlling the addition rate. After the hydrazine addition was complete, the ice bath was removed, and the reaction was allowed to stir at ambient temperature for 16 hours. The reaction was concentrated in vacuo and the residue dissolved in DCM and re-concentrated, then dried for 2 days to yield ethyl 1-methyl-1H-pyrazole-4-carboxylate (106 g, 99% yield) as a tan orange oil. MS (apci) m/z=155.1 (M+H).


Step B: 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile

To a four-necked 5-liter round bottomed flask fitted with an overhead stirrer and addition funnel was charged LHMDS (1444 mL, 1444 mmol) (1.0 M in THF). The solution was cooled in an acetone/dry ice bath first (internal temperature of −79° C.) under nitrogen, followed by slow addition of propiononitrile (103 mL, 1444 mmol) via dropping funnel. The mixture was stirred at −80° C. for 90 minutes. A solution of ethyl 1-methyl-1H-pyrazole-4-carboxylate (106 g, 688 mmol) in anhydrous THF (500 mL) was then introduced dropwise via an addition funnel (addition time: about 45 minutes; internal temperature during addition remained below −76° C.). After the addition was complete, the reaction was allowed to slowly warm to ambient temperature and stirred overnight. An orange glass deposited on the bottom of the flask. The organics were decanted and the glass was dissolved in warm water. The mixture was washed with ether (3×1000 mL). The aqueous phase was then pH-adjusted to 5 (pH paper) using concentrated HCl and saturated bicarbonate solution The aqueous layer was extracted with DCM (3×1000 mL). The combined organic extracts were dried over MgSO4 filtered and concentrated to yield 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile as an amber oil (92 g, 82% yield). MS (apci) m/z=162.1 (M−H).


Step C: 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine

A 3 L, 3 necked round bottomed flask was charged with 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile (60 g, 368 mmol) absolute anhydrous ethanol (1000 mL) and phenylhydrazine hydrochloride (58 g, 404 mmol) at ambient temperature to form a yellowish suspension. The reaction vessel was equipped with a water condenser and refluxed (using a heating mantle) overnight. The reaction was concentrated and 1M NaOH (1 L) was added and the solid was broken up and collected. The solid was washed with water and hexanes. A second crop crashed out in the filtrate and was collected. The combined solids were crushed and triturated with ether (500 mL). The solid was collected by filtration, washed with hexanes and dried in vacuo to provide the title compound (93 g, 100% yield) as a yellow solid. MS (apci) m/z=254.1 (M+H).


Intermediate 2



embedded image


phenyl (1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)carbamate

A 3 L, round bottomed flask was charged with 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine [Intermediate 1](50 g, 197.4 mmol) and EtOAc (1000 mL) to obtain a clear brownish solution. To this was added NaOH (2M aq) (500 mL) in one portion to obtain a turbid mixture (the aqueous and organic layers were clear, but a precipitate was observed in between the two layers). After 3 minutes, phenyl carbonochloridate (74.29 mL, 592.2 mmol) was added slowly at ambient temperature (the temperature of the reaction mixture increased to 33° C. during the addition). The reaction stirred at ambient temperature for 2 hours. Additional phenyl carbonochloridate (10 mL) was added. After 30 minutes the organics layers were separated, washed with brine and concentrated in vacuo. The residue was purified by silica column chromatography eluting with 75% EtOAc/hexanes to provide the title compound (60 g, 81% yield) as a cream foam. MS (apci) m/z=374.1 (M+H).


Intermediate 3



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl trifluoromethanesulfonate
Step A: Preparation of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one

A mixture of ethyl 2-cyanopropanoate (50.5 g, 397.2 mmol) and phenylhydrazine (39 mL, 397.2 mmol) in dioxane (100 mL) was heated at 110° C. for 5 days. The cooled mixture was concentrated to ½ volume then cooled in ice and triturated with cold Et2O. Solids were filtered, washed extensively with Et2O and dried in vacuo to afford 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (34.69 g, 46% yield) as a fluffy white powder. MS (apci) m/z=190.1 (M+H).


Step B: Preparation of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl trifluoromethane sulfonate

A suspension of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (13.72 g, 72.5 mmol) and N-phenylbis(trifluoromethylsulfonamide) (27.2 g, 76.1 mmol) in DMF (100 mL) was treated with DIEA (37.9 mL, 217.5 mmol) and the mixture stirred at ambient temperature for 16 hours. The mixture was partitioned between sat. NaHCO3 (400 mL) and EtOAc (200 mL) and the aqueous layer was extracted with EtOAc (2×200 mL). The combined organic phases were washed with water (5×50 mL) and brine (50 mL) then dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica column chromatography eluting with 4:1 hexanes/EtOAc, to afford the title compound (23.1 g, 99% yield) as a pale yellow solid. MS (apci) m/z=322.0 (M+H).


Intermediate 4



embedded image


3-bromo-4-methyl-1-phenyl-1H-pyrazol-5-amine

To a suspension of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one [Intermediate 3, step A](1.60 g, 8.46 mmol) in acetonitrile (30 mL) was added phosphorus oxybromide (3.64 g, 12.7 mmol) in one portion. The mixture was stirred at reflux for 3 hours then cooled and concentrated in vacuo. The residue was treated with DCM (50 mL) then sat. NaHCO3 (50 mL) was slowly added. The mixture was stirred for 30 minutes then the layers separated and the aqueous layer extracted with DCM (2×50 mL). The combined organic phases were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica column chromatography eluting with 2:1 hexanes/EtOAc, to afford the title compound (273 mg, 13% yield) as a white solid. MS (apci) m/z=254.0 (M+H).


Intermediate 5



embedded image


5-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-1-methylpyridin-2(1H)-one

3-Bromo-4-methyl-1-phenyl-1H-pyrazol-5-amine [Intermediate 4](763 mg, 3.03 mmol), 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2(1H)one (1.42 g, 6.05 mmol), K2CO3 (1.67 g, 12.1 mmol) and Pd(PPh3)4(350 mg, 0.30 mmol) were combined in toluene (10 mL), water (5 mL) and EtOH (2.5 mL) and warmed to 95° C. in a sealed tube for 16 hours. The cooled mixture was filtered and the filtrate partitioned between water (30 mL) and EtOAc (30 mL). The aqueous layer was extracted with EtOAc (2×20 mL) and the combined organic phases were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by silica column chromatography eluting with 2% MeOH/DCM to afford the title compound (504 mg, 59% yield) as a yellow foam. MS (apci) m/z=281.2 (M+H).


Intermediate 6



embedded image


phenyl (4-methyl-3-(1-methyl-6-oxo-1,6-dihydropyridin-3-yl)-1-phenyl-1H-pyrazol-5-yl)carbamate

To a suspension of 5-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-1-methylpyridin-2(1H)-one [Intermediate 5] (2.80 g, 9.99 mmol) in EtOAc (120 mL) was added 2N NaOH (14.98 mL, 29.97 mmol) followed by phenyl chloroformate (2.5 mL, 19.98 mmol). The mixture was stirred at ambient temperature for 16 hours then partitioned between water (100 mL) and EtOAc (100 mL) and the aqueous layer extracted with EtOAc (2×50 mL). The combined organic phases were washed with sat. NaHCO3 (50 mL) and brine (50 mL) then dried over Na2SO4, filtered and concentrated to afford the title compound as a pale yellow syrup which was used directly without purification, assuming 100% yield. MS (apci) m/z=401.2 (M+H).


Intermediate 7



embedded image


4-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-1-methylpyridin-2(1H)-one

Prepared according to the procedure of Intermediate 5, replacing 3-bromo-4-methyl-1-phenyl-1H-pyrazol-5-amine with 5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl trifluoromethanesulfonate [Intermediate 3] and 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2(1H)one with 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2(1H)-one. The product was purified by silica column chromatography eluting with 2% MeOH/DCM to afford the title compound (160 mg, 37% yield) as a pink solid. MS (apci) m/z=281.1 (M+H).


Intermediate 8



embedded image


phenyl (4-methyl-3-(1-methyl-2-oxo-1,2-dihydropyridin-4-yl)-1-phenyl-1H-pyrazol-5-yl)carbamate

Prepared according to the procedure of Intermediate 6, replacing 5-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-1-methylpyridin-2(1H)-one with 4-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-1-methylpyridin-2(1H)-one. MS (apci) m/z=401.1 (M+H).


Table 1 provides a list of commercially available pyrazole intermediates can be used in the synthesis of compounds described in the Examples.











TABLE 1





Pyrazole
Vendor/Catalog#
CAS#









embedded image


Oakwood, 021512
126208-61-5







embedded image


Array BioPharma, A1075-0
N/A







embedded image


Maybridge, GK03066
1192-21-8







embedded image


Ryan Scientific, EN300-14400
89399-92-8







embedded image


Oakwood, 021516
N/A







embedded image


Alfa Aesar, AAB20095-06
118430-73-2







embedded image


Aldrich, 532223
3524-32-1







embedded image


Accela ChemBio Chem Co, SY003755
876299-97-7







embedded image


ChemImpex, 18122
778611-16-8







embedded image


Oakwood, 017105
175137-45-8







embedded image


Alfa Aesar, AAB20464-06
5356-71-8







embedded image


Aldrich, 541001
1131-18-6







embedded image


Alfa Aesar, AAA15754-06
10199-50-5







embedded image


TCI America, A0174
826-85-7







embedded image


Oakwood, 023890
N/A







embedded image


J&W Pharmalab, 68-0035S
1187931-80-1







embedded image


VWR, EN300-09508
N/A







embedded image


ChemBridge, 4019184
885529-68-0







embedded image


ChemBridge, 4001950
N/A







embedded image


ChemImpex, 19156
337533-96-7







embedded image


Chemlmpex, 19155
898537-77-4







embedded image


ChemBridge, 4006072
N/A







embedded image


Oakwood, 005982
5346-56-5







embedded image


ChemImpex, 18771
182923-55-3







embedded image


Maybridge, KM00278
118430-74-3







embedded image


Maybridge, KM00835
118430-78-7







embedded image


ChemBridge, 4015288
N/A







embedded image


ChemBridge, 4015289
N/A







embedded image


Matrix, 020274
N/A







embedded image


Matrix, 019183
N/A







embedded image


Maybridge, KM 04038
126417-82-1







embedded image


ChemBridge, 4001950
N/A







embedded image


Lancaster, AAA17470-06
7152-40-1







embedded image


ChemBridge, 4010196
91642-97-6







embedded image


VWR, AAA13296-14
16078-71-0





N/A = Not available






Intermediate P1



embedded image


Ethyl 3-(5-amino-3-tert-butyl-1H-pyrazol-1-yl)benzoate

To a suspension of ethyl 3-hydrazinylbenzoate hydrochloride (500 mg, 2.31 mmol) in EtOH (20 mL) was added 4,4-dimethyl-3-oxopentanenitrile (318 mg, 2.54 mmol). The reaction mixture was heated to reflux for 18 hours, then cooled to ambient temperature and concentrated in vacuo. The crude product was purified by silica column chromatography, eluting with 0-5% MeOH/DCM to yield the product as a yellow oil (154 mg, 23% yield). MS (apci) m/z=288.2 (M+H).


The compounds in Table 2 were prepared by the method as described for Intermediate P1, substituting 4,4-dimethyl-3-oxopentanenitrile with the appropriate cyanoketone and ethyl 3-hydrazinylbenzoate hydrochloride with the appropriate hydrazine.











TABLE 2





Intermediate #
Structure
Data







P2


embedded image


MS (apci) m/z = 188.2 (M + H)





P3


embedded image


MS (apci) m/z = 218.1 (M + H)





P4


embedded image


MS (apci) m/z = 218.2 (M + H)





P5


embedded image


MS (apci) m/z = 188.2 (M + H)





P6


embedded image


MS (apci) m/z = 214.2 (M + H)





P7


embedded image


MS (apci) m/z = 188.2 (M + H)





P8


embedded image


MS (apci) m/z = 301.0 (M + H)





P9


embedded image


MS (apci) m/z = 218.1 (M + H)





P10


embedded image


MS (apci) m/z = 175.2 (M + H)





P11


embedded image


MS (apci) m/z = 237.3 (M + H)





P12


embedded image


MS (apci) m/z = 188.2 (M + H)





P13


embedded image


MS (apci) m/z = 188.2 (M + H)





P14


embedded image


MS (apci) m/z = 188.2 (M + H)





P15


embedded image


MS (apci) m/z = 204.2 (M + H)





P16


embedded image


MS (apci) m/z = 204.2 (M + H)





P17


embedded image


MS (apci) m/z = 199.0 (M + H)





P18


embedded image


MS (apci) m/z = 199.1 (M + H)





P19


embedded image


MS (apci) m/z = 192.2 (M + H)





P20


embedded image


MS (apci) m/z = 192.2 (M + H)





P21


embedded image


MS (apci) m/z = 232.2 (M + H)





P22


embedded image


MS (apci) m/z = 204.2 (M + H)





P23


embedded image


MS (apci) m/z = 206.1 (M + H)









Intermediate P101



embedded image


2-(1-methyl-1H-pyrazol-4-yl)-2,4,5,6-tetrahydrocyclopenta-[c]pyrazol-3-amine
Step A: Preparation of di-tert-butyl 1-(1-methyl-1H-pyrazol-4-yl)hydrazine-1,2-dicarboxylate

To a solution of 4-bromo-1-methyl-1H-pyrazole (1.93 mL, 18.6 mmol) in ether (37.3 mL) cooled to −78° C. was added nBuLi (23.3 mL, 37.3 mmol). After stirring at −78° C. for 30 minutes, a solution of di-t-butyl azodicarboxylate (4.29 g, 18.6 mmol) in Et2O (37.3 mL, 18.6 mmol) was added dropwise. After 1 hour, the reaction mixture was warmed up to −20° C. and quenched with ice. After warming to ambient temperature, the mixture was filtered and rinsed with Et2O. The resulting solid was taken up in a mixture of DCM and water, and the mixture was phase separated. The organic layer was dried with MgSO4, filtered and concentrated in vacuo to afford the first batch of product as a white solid (1.64 g, 28% yield). A second batch of product was recovered from the filtrate by silica column chromatography, eluting with 40-60% hexanes/EtOAc (0.51 g, 8.8% yield). MS (apci) m/z=313.0 (M+H).


Step B: Preparation of 2-(1-methyl-1H-pyrazol-4-yl)-2,4,5,6-tetrahydrocyclopenta-[c]pyrazol-3-amine

To a solution of di-tert-butyl 1-(1-methyl-1H-pyrazol-4-yl)hydrazine-1,2-dicarboxylate (103 mg, 0.330 mmol) in EtOH (1.65 mL, 0.330 mmol) was added concentrated HCl (137 μL, 1.65 mmol). The mixture was stirred at ambient temperature for 5 minutes, then cooled in an ice bath followed by addition of 2-oxocyclopentanecarbonitrile (36.0 mg, 0.330 mmol). After stirring for 5 minutes, the reaction mixture was warmed to ambient temperature overnight. The reaction mixture was concentrated and partitioned in water and DCM. After phase-separation, the aqueous layer was basified (pH 10) and then extracted with DCM (3×10 mL). The combined organic extracts were dried with MgSO4, filtered and concentrated in vacuo. The crude material was purified by reverse-phase column chromatography, eluting with 0-100% acetonitrile/water to afford the product as a yellow solid (4.5 mg, 6.7% yield). MS (apci) m/z=204.1 (M+H).


Intermediate P102



embedded image


3-tert-butyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-5-amine
Step A: Preparation of (tetrahydro-2H-pyran-4-yl)hydrazine hydrochloride

A suspension of dihydro-2H-pyran-4(3H)-one (2.00 g, 20.0 mmol) and tert-butyl hydrazinecarboxylate (2.64 g, 20.0 mmol) in hexanes (20.0 mL) was refluxed for 2 hours. After cooling, BH3-THF complex (20.0 mL, 20.0 mmol) was added and the reaction mixture was stirred for 1 hour. The mixture was then treated with 4 N HCl in dioxane (20.0 mL, 79.9 mmol), followed by 3 drops of water. After stirring at ambient temperature for 1 hour, the reaction mixture was filtered and rinsed with EtOAc to afford the product as a solid (2.39 g, 78.4% yield). MS (apci) m/z=117.0 (M+H).


Step B: Preparation of 3-tert-butyl-1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-5-amine

Prepared by the method as described in for the preparation of Intermediate P1, substituting (tetrahydro-2H-pyran-4-yl)hydrazine dihydrochloride for ethyl 3-hydrazinylbenzoate hydrochloride to yield the product as a yellow oil (0.472 g, 99.9% yield). MS (apci) m/z=224.1 (M+H).


Intermediate P103



embedded image


2-(pyridin-2-yl)-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine
Step A: Preparation of 2-(2-(pyridin-2-yl)hydrazono)cyclopentanecarbonitrile

A solution of 2-hydrazinylpyridine (0.200 g, 1.83 mmol) and 2-oxocyclopentanecarbonitrile (0.200 g, 1.83 mmol) in MeOH (9.16 mL) was treated with concentrated HCl (0.764 mL, 9.16 mmol) and refluxed for 16 hours. The reaction mixture was concentrated in vacuo, and then partitioned in water and DCM. After phase-separation, the aqueous layer was washed with DCM, basified (saturated NaHCO3, pH 10), and extracted with DCM. The combined organic layers were dried with MgSO4, filtered and concentrated. The crude material was purified by silica column chromatography, eluting with 100% EtOAc to afford the product (0.289 g, 78.6% yield). MS (apci) m/z=201.2 (M+H).


Step B: Preparation of 2-(pyridin-2-yl)-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine

A solution of 2-(2-(pyridin-2-yl)hydrazono)cyclopentanecarbonitrile (0.243 g, 1.21 mmol) in EtOH (6.06 mL, 1.21 mmol) was treated with 6 M HCl (0.202 mL, 1.21 mmol) and refluxed for 3 days. After removal of the solvent, the crude residue was diluted in water, basified (saturated NaHCO3, pH 10) and extracted with DCM. The combined organic layers were dried with MgSO4, filtered and concentrated. The crude material was purified by silica column chromatography, eluting with 50% EtOAc/hexanes to afford the product (0.198 g, 81.6% yield). MS (apci) m/z=201.2 (M+H).


Intermediate P104



embedded image


2-(pyridin-3-yl)-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine

Prepared by the method described above for Intermediate P103, substituting 3-hydrazinylpyridine for 2-hydrazinyl pyridine to afford the title product. MS (apci) m/z=201.1 (M+H).


Intermediate P105



embedded image


6,6-dimethyl-2-phenyl-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine
Step A: Preparation of 5-chloro-2,2-dimethylpentanenitrile

Isobutyronitrile (1.38 g, 20.0 mmol) and 1-bromo-3-chloropropane (3.46 g, 22.0 mmol) were sequentially added to a 1 M solution of lithium bis(trimethylsilyl)amide (20.0 mL, 20.0 mmol) while stirring. After stirring at 70° C. for 16 hours, the reaction mixture was quenched with water then extracted with DCM. The combined organic layers were dried with MgSO4, filtered and concentrated in vacuo to afford 5-chloro-2,2-dimethylpentanenitrile (2.91 g, 100% yield). 1H NMR (CDCl3) δ 3.57-3.61 (m, 2H), 1.94-2.02 (m, 2H), 1.67-1.72 (m, 2H), 1.37 (s, 6H).


Step B: Preparation of 2,2-dimethylhexanedinitrile

A suspension of 5-chloro-2,2-dimethylpentanenitrile (2.91 g, 20.0 mmol) and NaCN (1.57 g, 32.0 mmol) in DMF (20.0 mL) and water (1 mL) was heated at 100° C. for 16 hours. After cooling, the reaction mixture was diluted with water and refluxed for 30 minutes, then cooled, poured into water and stirred for 3 hours. The solution was then extracted with Et2O. The combined Et2O extracts were washed with H2O, dried with MgSO4, filtered and concentrated in vacuo to afford the product (2.20 g, 80.7% yield). 1H NMR (CDCl3) δ 2.42-2.47 (m, 2H), 1.83-1.92 (m, 2H), 1.67-1.72 (m, 2H), 1.39 (s, 6H).


Step C: Preparation of 3,3-dimethyl-2-oxocyclopentanecarbonitrile

A suspension of KOtBu (0.511 g, 4.55 mmol) in toluene (18.4 mL) was treated a toluene (2.0 mL) solution of 2,2-dimethylhexanedinitrile (1.00 g, 7.34 mmol) and heated at 80° C. for 2 hours. The reaction mixture was then cooled to ambient temperature and quenched with water. The mixture was separated and the organic layer was stirred in 2 N HCl (20 mL) for 16 hours. The mixture was separated and the organic layer dried with MgSO4, filtered and concentrated in vacuo to a yellow-white solid. The crude solid was purified by silica column chromatography, eluting with 10-40% EtOAc/hexanes, to afford the product (0.250 g, 24.8% yield). 1H NMR (CDCl3) δ 3.20-3.26 (m, 1H), 2.38-2.47 (m, 1H), 2.14-2.25 (m, 1H), 1.97-2.05 (m, 1H), 1.74-1.83 (m, 1H), 1.14 (s, 6H).


Step D: Preparation of 6,6-dimethyl-2-phenyl-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine

Prepared by the method as described for Intermediate P1, substituting phenylhydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 3,3-dimethyl-2-oxocyclopentanecarbonitrile for 4,4-dimethyl-3-oxopentanenitrile to afford the product (0.192 g, 46.2% yield) as a yellow solid. MS (apci) m/z=228.2 (M+H).


Intermediate P106



embedded image


7,7-dimethyl-2-phenyl-4,5,6,7-tetrahydro-2H-indazol-3-amine
Step A: Preparation of 2,2-dimethylheptanedinitrile

Prepared by the method as described for Intermediate P105, Steps A and B, substituting 1-bromo-4-chlorobutane for 1-bromo-3-chloropropane to yield the product (2.21 g, 73.7% yield). 1H NMR (CDCl3) δ 2.37-2.42 (m, 2H), 1.53-1.77 (m, 6H), 1.36 (s, 6H).


Step B: Preparation of 3,3-dimethyl-2-oxocyclohexanecarbonitrile

A suspension of KOtBu (0.463 g, 4.13 mmol) in toluene (16.6 mL) was treated with a solution of 2,2-dimethylheptanedinitrile (1.00 g, 6.66 mmol) in toluene (2.0 mL) and heated at 80° C. for 48 hours. After cooling to ambient temperature, the reaction mixture was quenched with water and phase-separated, and the organic layer was stirred with 2 N HCl (20 mL) for 16 hours. After phase-separation, the organic layer was dried with MgSO4, filtered and concentrated in vacuo. The crude material was purified by silica column chromatography, eluting with 10-20% EtOAc/hexanes to afford the product (0.374 g, 37.2% yield). 1H NMR (CDCl3) δ 3.72-3.78 (m, 1H), 2.42-2.50 (m. 1H), 1.78-2.04 (m, 4H), 1.60-1.70 (m, 1H), 1.21 (s, 3H), 1.16 (s, 3H).


Step C: Preparation of 7,7-dimethyl-2-phenyl-4,5,6,7-tetrahydro-2H-indazol-3-amine

Prepared by the method as described for Intermediate P1, substituting phenylhydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 3,3-dimethyl-2-oxocyclohexanecarbonitrile for 4,4-dimethyl-3-oxopentanenitrile to yield the product as an off-white solid (0.490 g, 54.2% yield, 66% purity). MS (apci) m/z=242.2 (M+H).


Intermediate P107



embedded image


3-isopropyl-4-methyl-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of 2,4-dimethyl-3-oxopentanenitrile

To a solution of propiononitrile (518 mg, 9.40 mmol) in THF (50 mL, 7.83 mmol) at −78° C. under N2 was slowly added lithium bis(trimethylsilyl)amide (1M in THF) (7.83 mL, 7.83 mmol). After 30 minutes, methyl isobutyrate (0.898 mL, 7.83 mmol) was added dropwise, and the reaction mixture was warmed to 0° C. A yellow precipitate formed, the reaction mixture was stirred for 1 hour, then diluted with H2O (50 mL) to dissolve the solids. The mixture was extracted with Et2O (25 mL), and the basic aqueous phase was acidified with 2M HCl (5 mL) and extracted with Et2O (2×50 mL). The combined organic phases were washed with brine (50 mL), dried with MgSO4, filtered, and concentrated to afford the product (421 mg, 42.9% yield)


Step B: Preparation of 3-isopropyl-4-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P1, substituting phenyl hydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 4,4-dimethyl-3-oxopentanenitrile with 2,4-dimethyl-3-oxopentanenitrile to yield the product as a yellow syrup (0.587 g, 81.1% yield). MS (apci) m/z=216.2 (M+H).


Intermediate P108



embedded image


2-phenyl-4,6-dihydro-2H-furo [3,4-c]pyrazol-3-amine
Step A: Preparation of 4-oxotetrahydrofuran-3-carbonitrile

To a suspension of KOtBu (996.6 mg, 8.881 mmol) in THF (640.4 mg, 8.881 mmol) cooled to 0° C. was added dropwise methyl 2-hydroxyacetate (675.7 μL, 8.881 mmol) and stirred for 10 minutes. The acrylonitrile (589.1 μL, 8.881 mmol) was then added and the reaction stirred at ambient temperature. After 3 hours, the reaction was diluted with H2O (50 mL), then extracted with Et2O (25 mL) to remove any starting ester. The basic aqueous phase was acidified with 2M HCl (5 mL), then extracted with Et2O (2×50 mL). The combined organic phases were dried with MgSO4, filtered, and concentrated to afford a light brown oil (446 mg, 45.2% yield). 1H NMR (CDCl3) δ 4.63 (t, 1H), 4.24 (t, 1H), 4.14 (d, 1H), 4.02 (d, 1H), 3.57 (t, 1H).


Step B: Preparation of 2-phenyl-4,6-dihydro-2H-furo[3,4-c]pyrazol-3-amine

Prepared by the method as described for Intermediate P1, substituting phenyl hydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 4,4-dimethyl-3-oxopentanenitrile with 4-oxotetrahydrofuran-3-carbonitrile to yield the product as a reddish-brown syrup (182 mg, 22.5% yield). MS (apci) m/z=202.1 (M+H).


Intermediate P109



embedded image


3-(methoxymethyl)-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of 4-methoxy-3-oxobutanenitrile

To a solution of methyl 2-methoxyacetate (0.4753 mL, 4.803 mmol) in THF (20 mL, 4.803 mmol) at −78° C. under N2 was added acetonitrile (0.3033 mL, 5.763 mmol), followed by lithium bis(trimethylsilyl)amide (1M in THF) (4.803 mL, 4.803 mmol). After stirring 1 hour, the reaction mixture was warmed to 0° C. and stirred for 1 hour. The reaction mixture was then diluted with H2O (25 mL), washed with Et2O (25 mL), then neutralized with 2 M HCl (1.5 mL). This was extracted with Et2O (2×25 mL) and the combined organic phases were washed with brine (25 mL), dried with MgSO4, filtered, and concentrated to afford the product (169 mg, 31.1% yield). 1H NMR (CDCl3) δ 4.09 (s, 2H), 3.66 (s, 2H), 3.46 (s, 3H)


Step B: Preparation of 3-(methoxymethyl)-1-phenyl-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P1, substituting phenyl hydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 4,4-dimethyl-3-oxopentanenitrile with 4-methoxy-3-oxobutanenitrile to yield the product as a pale yellow residue (6.0 mg, 2.0% yield). MS (apci) m/z=204.0 (M+H).


Intermediate P110



embedded image


3-(methoxymethyl)-4-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared according to the method as described for Intermediate P109, replacing acetonitrile with propionitrile to afford the product as an orange residue. MS (apci) m/z=218.0 (M+H).


Intermediate P111



embedded image


2-(5-amino-1-phenyl-1H-pyrazol-3-yl)-2-methylpropan-1-ol
Step A: Preparation of methyl 3-(tert-butyldimethylsilyloxy)-2,2-dimethylpropanoate

Methyl 3-hydroxy-2,2-dimethylpropanoate (1.000 g, 7.567 mmol), TBDMS-Cl (1.140 g, 7.567 mmol) and imidazole (0.5666 g, 8.323 mmol) were dissolved in DMF (5 mL, 7.567 mmol) and stirred at ambient temperature overnight. The reaction mixture was diluted with H2O (25 mL) and extracted with EtOAc (2×25 mL). The combined organic phases were washed with brine (25 mL), dried with MgSO4, filtered and concentrated to afford the product (1.92 g, 103% yield). 1H NMR (CDCl3) δ 3.66 (s, 3H), 3.57 (s, 2H), 1.15 (s, 6H), 0.87 (s, 9H), 0.02 (s, 6H).


Step B: Preparation of 5-(tert-butyldimethylsilyloxy)-4,4-dimethyl-3-oxopentanenitrile

Prepared according to the method described for Intermediate P109, replacing methyl 2-methoxyacetate with methyl 3-(tert-butyldimethylsilyloxy)-2,2-dimethylpropanoate to afford the product as a pale yellow residue. 1H NMR (CDCl3) δ 3.70 (s, 2H), 3.55 (s, 2H), 1.15 (s, 6H), 0.89 (s, 9H), 0.06 (s, 6H).


Step C: Preparation of 2-(5-amino-1-phenyl-1H-pyrazol-3-yl)-2-methylpropan-1-ol

Prepared by the method as described for Intermediate P1, substituting phenyl hydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 4,4-dimethyl-3-oxopentanenitrile with methyl 3-(tert-butyldimethylsilyloxy)-2,2-dimethylpropanoate to yield the product as yellow syrup (74 mg, 66% yield). MS (apci) m/z=232.2 (M+H).


Intermediate P112



embedded image


2-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-2-methylpropan-1-ol

Prepared according to the method described for Intermediate P111, replacing acetonitrile with propionitrile to afford the product as a yellow residue. MS (apci) m/z=246.2 (M+H).


Intermediate P113



embedded image


3-(3-methoxypropyl)-4-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared according to the method described for Intermediate P109, replacing methyl 2-methoxyacetate with methyl 4-methoxybutanoate and replacing acetonitrile with propionitrile in Step A to afford the product as an orange-brown syrup. MS (apci) m/z=246.1 (M+H).


Intermediate P114



embedded image


1,1′-dimethyl-1H,1′H-3,4′-bipyrazol-5-amine
Step A: Preparation of 3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile

A solution of ethyl 1-methyl-1H-pyrazole-4-carboxylate (500 mg, 3.24 mmol), toluene (7.50 mL, 70.4 mmol), and acetonitrile (346 μL, 6.49 mmol) was treated in one portion with KOtBu (1092 mg, 9.73 mmol) to give a hazy solution. The reaction was allowed to stir at ambient temperature for one hour, and was determined to be complete by HPLC analysis. The mixture was treated with water (7.5 mL) and stirred for 1 minute, then acidified with 3M HCl (3027 μL, 9.08 mmol) to pH 5.5-6. The aqueous layer was extracted with ethyl acetate (3×5 mL) and the combined organic extracts were concentrated in vacuo to give a yellow viscous oil, which completely solidified upon placing under high vacuum to afford the product (102 mg, 21.1% yield). 1H NMR (CDCl3) δ 8.02 (s, 1H), 7.94 (s, 1H), 3.98 (s, 3H), 3.82 (s, 2H)


Step B: Preparation of 1,1′-dimethyl-1H,1′H-3,4′-bipyrazol-5-amine

Prepared by the method as described for Intermediate P1, substituting methyl hydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and replacing 4,4-dimethyl-3-oxopentanenitrile with 3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile to yield the product as an ivory white solid (45 mg, 44.6% yield). MS (apci) m/z=178.1 (M+H).


Intermediate P115



embedded image


4-chloro-1,3-diphenyl-1H-pyrazol-5-amine

To a solution of 1,3-diphenyl-1H-pyrazol-5-amine (Table 1; 0.100 g, 0.425 mmol) in acetonitrile (2 mL) was added N-chlorosuccinimide (0.0568 g, 0.425 mmol). The pale yellow solution was stirred at ambient temperature for 3 hours, then concentrated in vacuo and purified by silica column chromatography eluting with 20% EtOAc/Hexanes to afford the product as a light brown oil (0.10 g, 87% yield). MS (apci) m/z=270.0 (M+H).


Intermediate P116



embedded image


4-bromo-1,3-diphenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate P115, substituting N-chloro succinimide with N-bromo-succinimide. MS (apci) m/z=313.9 (M+H).


Intermediate P117



embedded image


4-chloro-3-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate P115, substituting 1,3-diphenyl-1H-pyrazol-5-amine with 3-methyl-1-phenyl-1H-pyrazol-5-amine.


MS (apci) m/z=207.9 (M+H).


Intermediate P118



embedded image


4-bromo-3-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate P117, substituting N-chloro succinimide with N-bromo-succinimide. MS (apci) m/z=251.9 (M+H).


Intermediate P119



embedded image


4-chloro-1-methyl-3-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate P115, substituting 1,3-diphenyl-1H-pyrazol-5-amine with 1-methyl-3-phenyl-1H-pyrazol-5-amine (Table 1). MS (apci) m/z=208.0 (M+H).


Intermediate P120



embedded image


4-bromo-1-methyl-3-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate P119, substituting N-chloro succinimide with N-bromo-succinimide. MS (apci) m/z=251.9 (M+H).


Intermediate P121



embedded image


1-methyl-3-(4-(methylthio)phenyl)-1H-pyrazol-5-amine
Step A: Preparation of 3-(4-(methylthio)phenyl)-3-oxopropanenitrile

To a suspension of NaH (60% in mineral oil) (154 mg, 3.84 mmol) in dioxane (25.0 mL, 2.74 mmol) was added acetonitrile (0.217 mL, 4.12 mmol). The reaction mixture was stirred at ambient temperature for 30 minutes, then treated with methyl 4-(methylthio)benzoate (500 mg, 2.74 mmol) and heated to reflux for 15 hours. The suspension was cooled, then diluted with water (25 mL) and washed with Et2O (25 mL). The aqueous layer was neutralized with 2M HCl (1.8 mL) and extracted with Et2O (2×25 mL). The combined organic phases were washed with brine (25 mL), dried with MgSO4, filtered and concentrated in vacuo. The resultant residue was purified by silica column chromatography eluting with 0-5% MeOH/DCM to afford the product (317 mg, 60.4% yield). 1H NMR (CDCl3) δ 7.82 (d, 2H), 7.30 (d, 2H), 4.02 (s, 2H), 2.54 (s, 3H).


Step B: Preparation of 1-methyl-3-(4-(methylthio)phenyl)-1H-pyrazol-5-amine

Prepared by the method as described in Intermediate P1, substituting methylhydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and substituting 3-(4-(methylthio)phenyl)-3-oxopropanenitrile for 4,4-dimethyl-3-oxopentanenitrile to yield the product as a yellow solid (0.307 g, 96.7% yield). MS (apci) m/z=220.0 (M+H).


Intermediate P122



embedded image


2-(5-amino-1-phenyl-1H-pyrazol-3-yl)-2-methylpropanenitrile

Prepared according to the procedure for Intermediate P121, substituting methyl 4-(methylthio)benzoate with ethyl 2-cyano-2-methylpropanoate in Step A and phenyl hydrazine hydrochloride for methyl hydrazine in Step B. MS (apci) m/z=227.1 (M+H).


Intermediate P123



embedded image


3-(4-(2-methoxyethoxy)phenyl)-1-methyl-1H-pyrazol-5-amine
Step A: Preparation of 3-(4-(benzyloxy)phenyl)-3-oxopropanenitrile

Prepared according to the procedure described for Intermediate P121, substituting methyl 4-(methylthio)benzoate with methyl 4-(benzyloxy)benzoate in Step A. 1H NMR (CDCl3) δ 7.90 (d, 2H), 7.42 (m, 4H), 7.37 (m, 1H), 7.05 (d, 2H), 5.16 (s, 2H), 4.00 (s, 2H).


Step B: Preparation of 3-(4-(benzyloxy)phenyl)-1-methyl-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P1, substituting methylhydrazine for ethyl 3-hydrazinylbenzoate hydrochloride and 3-(4-(benzyloxy)phenyl)-3-oxopropanenitrile for 4,4-dimethyl-3-oxopentanenitrile to yield the product as a yellow solid. MS (apci) m/z=280.1 (M+H).


Step C: Preparation of 4-(5-amino-1-methyl-1H-pyrazol-3-yl)phenol

To a solution of 3-(4-(benzyloxy)phenyl)-1-methyl-1H-pyrazol-5-amine (47 mg, 0.17 mmol) in EtOH (5.0 mL) was added 5% Pd/C (9.0 mg, 0.0084 mmol) and stirred under a H2 balloon for 17 hours. The reaction mixture was filtered through Celite®, rinsed with EtOH and concentrated in vacuo to afford the product (28 mg, 88% yield). MS (apci) m/z=190.1 (M+H).


Step D: Preparation of 3-(4-(2-methoxyethoxy)phenyl)-1-methyl-1H-pyrazol-5-amine

To a solution of 4-(5-amino-1-methyl-1H-pyrazol-3-yl)phenol (14 mg, 0.074 mmol) in DMSO (0.50 mL, 7.0 mmol) was added Cs2CO3 (48 mg, 0.15 mmol) and 1-bromo-2-methoxyethane (9.7 μL, 0.10 mmol). The reaction mixture was stirred for 16 hours, then diluted with water (10 mL) and extracted with DCM (3×10 mL). The combined organic extracts were washed with brine (10 mL), dried with MgSO4, filtered and concentrated to afford the crude product (22 mg, 120% yield). The crude product was used without purification in subsequent steps. MS (apci) m/z=248.0 (M+H).


Intermediate P124



embedded image


1′-methyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine

Prepared according to the procedure described for Intermediate P114, substituting methylhydrazine with phenylhydrazine in Step B. MS (apci) m/z=240.0 (M+H).


Intermediate P125



embedded image


4-methoxy-3-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure for Intermediate P121, substituting methyl 4-(methylthio)benzoate with ethyl acetate and substituting acetonitrile with 2-methoxyacetonitrile in Step A and phenyl hydrazine hydrochloride for methyl hydrazine in Step B. MS (apci) m/z=204.0 (M+H).


Intermediate P126



embedded image


(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)methanol

Prepared according to the procedure for Intermediate P112, substituting methyl 3-hydroxy-2,2-dimethylpropanoate with ethyl 2-hydroxyacetate in Step A. MS (apci) m/z=204.1 (M+H).


Intermediate P127



embedded image


2-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)ethanol

Prepared according to the procedure for Intermediate P112, substituting methyl 3-hydroxy-2,2-dimethylpropanoate with methyl 3-hydroxypropanoate in Step A. MS (apci) m/z=218.0 (M+H).


Intermediate P128



embedded image


3-(2-methoxyethyl)-4-methyl-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of 5-methoxy-2-methyl-3-oxopentanenitrile

To a suspension of NaNH2 (50 wt % suspension in toluene) (330 mg, 4.23 mmol) in THF (25 mL, 4.23 mmol) under N2 at −78° C. was added propiononitrile (0.448 mL, 6.35 mmol), and the reaction mixture was stirred for 30 minutes. Methyl 3-methoxypropanoate (0.495 mL, 4.23 mmol) was added and the reaction mixture was stirred at −78° C. for 1 hour, then at 0° C. for 2.5 hours. The reaction mixture was diluted with H2O (25 mL) and washed with Et2O (25 mL). The basic aqueous phase was neutralized with 2M HCl (1.6 mL), then extracted with Et2O (3×25 mL). The combined organic phases were washed with brine (25 mL), dried with MgSO4, filtered, and concentrated to afford the crude product as a pale greenish oil (171 mg). The crude mixture was taken directly to the next step.


Step B: Preparation of 3-(2-methoxyethyl)-4-methyl-1-phenyl-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P1, substituting 5-methoxy-2-methyl-3-oxopentanenitrile for 4,4-dimethyl-3-oxopentanenitrile and substituting phenylhydrazine hydrochloride for ethyl 3-hydrazinylbenzoate hydrochloride to yield the product as a yellow solid (56 mg, 20% yield). MS (apci) m/z=232.0 (M+H).


Intermediate P129



embedded image


Phenyl (5-oxido-2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-yl)carbamate

A THF (4 mL) solution of phenyl 2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-ylcarbamate (Intermediate P130, Step B; 50 mg, 0.15 mmol) was cooled to −50° C. with an external dry-ice/MeCN bath and treated with a THF (2 mL) solution of 3-chlorobenzoperoxoic acid (33 mg, 0.13 mmol). After stirring for 1 hour, the mixture was quenched with Na2S2O3 and water, extracted with EtOAc, washed with NaHCO3 and brine, dried with MgSO4, filtered, and concentrated to give the product which was directly used in next step without further purification. MS (apci) m/z=354.1 (M+H).


Intermediate P130



embedded image


Phenyl (5,5-dioxido-2-phenyl-4,6-dihydro-2H-thieno [3,4-c]pyrazol-3-yl)carbamate
Step A: Preparation of 2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-amine

A suspension of 4-oxotetrahydrothiophene-3-carbonitrile (1.00 g, 7.86 mmol) and phenylhydrazine hydrochloride (1.25 g, 8.65 mmol) in absolute EtOH (40 mL) was refluxed for 2 hours. After removal of solvent under reduced pressure, the white solid residue was triturated with 1 N NaOH (40 mL). The solid was collected by filtration, washed with 0.1 N NaOH, water, and hexanes (approx. 10 mL each) then dried on high vacuum to yield the product as white solid (1.6 g, 95% yield). MS (apci pos) m/z=218.1 (M+H).


Step B: Preparation of phenyl 2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-ylcarbamate

To a suspension of 2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-amine (500 mg, 2.30 mmol) in EtOAc (10 mL) was added NaOH (2M aq, 2.3 mL, 4.60 mmol), followed by dropwise addition of phenyl carbonochloridate (0.400 mL, 3.22 mmol). After stirring at ambient temperature for 2 hours, another portion of phenyl carbonochloridate (0.16 mL, 1.3 mmol) was added dropwise, and the reaction was stirred at ambient temperature for 15 hours. The reaction mixture was diluted with EtOAc (20 mL) and phase-separated. The organic phase was washed with H2O, brine (25 mL each), then dried with Na2SO4, filtered and concentrated. The crude material was purified by reverse-phase column chromatography, eluting with 5-70% acetonitrile/water to yield the product as white solid (0.5 g, 64% yield). MS (apci pos) m/z=338.1 (M+H).


Step C: Preparation of phenyl (5,5-dioxido-2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-yl)carbamate

To a turbid solution of phenyl 2-phenyl-4,6-dihydro-2H-thieno[3,4-c]pyrazol-3-ylcarbamate (50 mg, 0.15 mmol) in DCM (1.5 mL) at 0° C. was added MCPBA (91 mg, 0.37 mmol, 70-75% water complex), and the mixture was stirred at ambient temperature for 10 min. The mixture was then diluted with DCM (3 mL) and washed with saturated aqueous NaHCO3 (3×2 mL) and saturated aqueous Na2S2O3 (3×2 mL). The organic layer was dried with MgSO4, filtered and concentrated under reduced pressure to yield the title product as light yellowish foamy solid (31 mg, 57% yield, 95% pure). MS (apci pos) m/z=371.0 (M+H).


Intermediate P132



embedded image


1-methyl-3-(pyrazin-2-yl)-1H-pyrazol-5-amine
Step A: Preparation of 3-oxo-3-(pyrazin-2-yl)propanenitrile

To a suspension of NaH (60% in mineral oil, 81.1 mg, 2.03 mmol) in dioxane (15 mL) was added acetonitrile (0.114 mL, 2.17 mmol), followed by methyl pyrazine-2-carboxylate (200 mg, 1.45 mmol) and the reaction heated to reflux for 2.5 hours. The reaction mixture was cooled to ambient temperature and diluted with H2O (25 mL) and extracted with Et2O (25 mL). The aqueous phase was neutralized with 2M aqueous HCl (0.7 mL), then extracted with 10% MeOH/DCM (3×25 mL). The combined organic phases were washed with brine (25 mL), dried with MgSO4, filtered, and concentrated to yield the crude product as an orange syrup (134 mg, 62.9% yield). 1H NMR (CDCl3) δ 9.32 (d, 1H), 8.87 (d, 1H), 8.68 (dd, 1H), 4.34 (s, 2H).


Step B: Preparation of 1-methyl-3-(pyrazin-2-yl)-1H-pyrazol-5-amine

To a suspension of 3-oxo-3-(pyrazin-2-yl)propanenitrile (67.0 mg, 0.455 mmol) in EtOH (5 mL) was added methylhydrazine (0.024 mL, 0.455 mmol). The reaction mixture was refluxed for 15 hours, then concentrated in vacuo. The crude product was purified by silica column chromatography, eluting with 0-5% MeOH/DCM to yield the product as a brown residue (33 mg, 41% yield). MS (apci) m/z=176.2 (M+H).


Intermediate P133



embedded image


1-methyl-3-(5-methylpyrazin-2-yl)-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P107, substituting methyl isobutyrate in Step A with methyl 5-methylpyrazine-2-carboxylate and propionitrile with acetonitrile to afford 3-(5-methylpyrazin-2-yl)-3-oxopropanenitrile. In Step B, phenylhydrazine was replaced by methylhydrazine to afford the title pyrazole. MS (apci) m/z=190.2 (M+H).


Intermediate P134



embedded image


1,4-dimethyl-3-(5-methylpyrazin-2-yl)-1H-pyrazol-5-amine

Prepared by the method as described for Intermediate P107, substituting methyl isobutyrate in Step A with methyl 5-methylpyrazine-2-carboxylate to afford 2-methyl-3-(5-methylpyrazin-2-yl)-3-oxopropanenitrile. In Step B, phenylhydrazine was replaced by methylhydrazine to afford the title compound. MS (apci) m/z=204.1 (M+H).


Intermediate P135



embedded image


3-ethoxy-4-methyl-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one

A mixture of ethyl 2-cyanopropanoate (5.0 g, 46 mmol) and phenylhydrazine (5.9 g, 46 mmol) in dioxane (10 mL) was heated at 110° C. for 17 hours. The crude material was cooled to ambient temperature, concentrated, and triturated with cold EtOH and Et2O. The resultant solid was filtered, washed with Et2O, and dried under vacuum to give the product as a white solid (3.4 g, 39% yield). MS (apci) m/z=190.0 (M−H).


Step B: Preparation of 3-ethoxy-4-methyl-1-phenyl-1H-pyrazol-5-amine

To a suspension of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (10.0 g, 52.9 mmol) in DMF (100 mL) was added K2CO3 (14.6 g, 106 mmol) and bromoethane (4.34 mL, 58.1) at ambient temperature. After stirring for 17 hours, the reaction mixture was treated with EtOAc and washed with water (3×, to obtain the N-alkylation product) and brine, dried with MgSO4, filtered, and concentrated to give the product (5.35 g, 47% yield). MS (apci) m/z=218.1 (M+H).


The compounds in Table 3 were prepared by the method as described for Intermediate P135, substituting bromoethane with the appropriate alkyl halide or alkyl methanesulfonate.











TABLE 3





Intermediate #
Structure
Data







P200


embedded image


MS (apci) m/z = 248.1 (M + H)





P201


embedded image


MS (apci) m/z = 204.1 (M + H)





P202


embedded image


MS (apci) m/z = 229.0 (M + H)





P203


embedded image


MS (apci) m/z = 348.1 (M + H)





P204


embedded image


MS (apci) m/z = 310.0 (M + H)





P205


embedded image


MS (apci) m/z = 236.1 (M + H)





P206


embedded image


MS (apci) m/z = 264.0 (M + H)





P207


embedded image


MS (apci) m/z = 260.1 (M + H)





P208


embedded image


MS (apci) m/z = 274.1 (M + H)





P209


embedded image


MS (apci) m/z = 304.1 (M + H)





P210


embedded image


MS (apci) m/z = 262.1 (M + H)





P211


embedded image


MS (apci) m/z = 362.0 (M + H)





P212


embedded image


MS (apci) m/z = 304.1 (M + H)









Intermediate P136



embedded image


3-(benzyloxy)-1-methyl-1H-pyrazol-5-amine
Step A: Preparation of 5-amino-1-methyl-4-phenyl-1H-pyrazol-3(2H)-one

To a suspension of ethyl 2-cyano-2-phenylacetate (2.56 g, 13.3 mmol) in EtOH (10 mL) was added dropwise methylhydrazine (1.09 mL, 19.9 mmol). The reaction was heated at 85° C. for 15 hours. The reaction mixture was cooled to 0° C. and filtered. The resultant solid was washed with cold EtOH (20 mL) and Et2O (20 mL) to give the desired product (2.10 g, 83.7% yield). MS (apci) m/z=190.2 (M+H)


Step B: Preparation of 3-(benzyloxy)-1-methyl-1H-pyrazol-5-amine

A suspension of 5-amino-1-methyl-1H-pyrazol-3(2H)-one (0.35 g, 3.1 mmol), Benzyl chloride (0.43 g, 3.4 mmol), and K2CO3 (1.3 g, 9.3 mmol) in DMF (4 mL) was heated at 70° C. for 17 hours. After cooling, the reaction mixture was treated with EtOAc, washed with water and brine, dried with MgSO4, and concentrated in vacuo. The crude product was purified by silica column chromatography eluting with 2-6% MeOH/DCM to afford the title compound (0.16 g, 25% yield). MS (apci) m/z=204.0 (M+H).


Intermediate P137



embedded image


3-methoxy-1-methyl-4-phenyl-1H-pyrazol-5-amine

To a suspension of 5-amino-1-methyl-4-phenyl-1H-pyrazol-3(2H)-one (Step A of the preparation of Intermediate P136; 208 mg, 1.10 mmol) and K2CO3 (456 mg, 3.30 mmol) in DMF (5 mL) was added dropwise iodomethane (172 mg, 1.21 mmol). The reaction mixture was stirred for 15 hours. The solvent was removed under reduced pressure and the residue was purified by silica column chromatography eluting with 33% EtOAc/Hexanes to give the title pyrazole (66.0 mg, 30.4% yield). MS (apci) m/z=204.1 (M+H).


Intermediate P138



embedded image


3-ethoxy-1-methyl-4-phenyl-1H-pyrazol-5-amine

Prepared as described in Intermediate P137, replacing iodomethane with iodoethane in Step B to afford the title compound. MS (apci) m/z=218.2 (M+H).


Intermediate P139



embedded image


3-ethoxy-1-phenyl-1H-pyrazol-5-amine

Prepared according to the procedure described for Intermediate 135, substituting ethyl-2-cyanopropanoate with ethyl-2-cyanoacetate in Step A. MS (apci) m/z=204.0 (M+H).


The compounds in the following Table were prepared by the method as described for Intermediate P135, substituting bromoethane with the appropriate alkyl halide, alkyl methanesulfonate or epoxide.














Intermediate #
Structure
MS (apci) m/z







P140


embedded image


286.1 (M + H)





P141


embedded image


303.1 (M + H)





P142


embedded image


262.1 (M + H)





P143


embedded image


402.2 (M + H)





P144


embedded image


276.1 (M + H)





P145


embedded image


363.1 (M + H)





P146


embedded image


248.1 (M + H)





P147


embedded image


248.1 (M + H)





P148


embedded image


302.1 (M + H)





P149


embedded image


302.1 (M + H)





P150


embedded image


262.1 (M + H)









Intermediate 151



embedded image


1′-(2-methoxyethyl)-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-amine
Step A: Preparation of methyl 1-methyl-1H-1,2,4-triazole-3-carboxylate

To a stirred suspension of NaH (60% oil dispersion, 0.346 g, 8.66 mmol) in DMF (20 mL) was added dropwise a solution of methyl 1H-1,2,4-triazole-3-carboxylate (1.00 g, 7.87 mmol) in DMF (20 mL) at 0° C. under nitrogen. The reaction mixture was stirred at 0° C. for 1 hour. MeI (0.982 mL, 15.7 mmol) was added dropwise. The reaction mixture was stirred at ambient temperature overnight. The reaction was poured into cold water and extracted with EtOAc. The combined organic layers were washed with brine, dried and concentrated. The residue was purified by column chromatography (3:1 hexanes/EtOAc) to give the title compound (0.380 g, 34% yield) as a white solid. MS (apci) m/z=142.1 (M+H).


Step B: Preparation of 1′-(2-methoxyethyl)-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-amine

Prepared according to the method described for Intermediate P109, using methyl 1-methyl-1H-1,2,4-triazole-3-carboxylate as a replacement for methyl 2-methoxyacetate, and substituting propionitrile for acetonitrile in Step A. MS (apci) m/z=255.1 (M+H).


Intermediate 152



embedded image


1′-(2-methoxyethyl)-4-methyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-amine

Prepared according to the method described for Intermediate P109, using ethyl 1-(2-methoxyethyl)-1H-pyrazole-4-carboxylate as a replacement for methyl 2-methoxyacetate, and substituting propionitrile for acetonitrile in Step A.


Intermediate 153



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbonitrile

To a stirred solution of aniline (2.02 g, 21.7 mmol) in 6 N HCl (22 mL) was added dropwise a solution of NaNO2 (1.50 g, 21.7 mmol) in water (20 mL) at 0-5° C. The reaction mixture was stirred at 0° C. for 15 minutes. Acetic acid (10 mL) was added. This solution was added dropwise to a stirred solution of ethyl 2,3-dicyanobutanoate (Prepared according to the procedure described in Bioorganic & Medicinal Chemistry, 2004, 12, 3345-3356, 3.60 g, 21.7 mmol) in acetic acid (12 mL) and water (18 mL) at 0° C. After stirring for 1 hour, concentrated ammonium hydroxide (50 mL) was added dropwise followed by THF (50 mL). The reaction was stirred at ambient temperature overnight. The organic layer was separated. The aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried and concentrated. The residue was purified by flash chromatography on silica gel (3:1 hexanes/EtOAc) to give the title compound (2.95 g, 69% yield). MS (apci) m/z=198.9 (M+H).


Intermediate 155



embedded image


4-methyl-3-(2-methyl-2H-1,2,3-triazol-4-yl)-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of ethyl 2-methyl-2H-1,2,3-triazole-4-carboxylate

A mixture of ethyl 2H-1,2,3-triazole-4-carboxylate (2.00 g, 14.2 mmol), K2CO3 (3.53 g, 25.5 mmol) and methyl iodide (3.54 mL, 56.7 mmol) in acetonitrile (40 mL) was stirred at 50° C. under nitrogen overnight. After cooling to ambient temperature, the mixture was filtered through Celite®. The filtrate was concentrated in vacuo. The residue was purified by flash chromatography on silica gel (4:1 hexane/EtOAc) to give the title compound (0.780 g, 35% yield). MS (apci) m/z=156.0 (M+H).


Step B: Preparation of 4-methyl-3-(2-methyl-2H-1,2,3-triazol-4-yl)-1-phenyl-1H-pyrazol-5-amine

Prepared according to the method described for Intermediate P109 using ethyl 2-methyl-2H-1,2,3-triazole-4-carboxylate as a replacement for methyl 2-methoxyacetate, and substituting propionitrile for acetonitrile in Step A. MS (apci) m/z=254.9 (M+H).


Intermediate 156



embedded image


3-bromo-4-methyl-1-phenyl-1H-pyrazol-5-amine

To a stirred solution of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (Intermediate P135, Step A, 1.00 g, 5.29 mmol) in MeCN (20 mL) was added POBr3 (2.27 g, 7.93 mmol). The reaction mixture was heated at reflux for 3 hours. The reaction was concentrate in vacuo. The residue was taken up in DCM. Saturated aqueous NaHCO3 solution was carefully added. The aqueous layer was extracted with DCM. The combined organic layers were washed with brine, dried and concentrated. The residue was purified by flash chromatography on silica gel (1:2 hexane/EtOAc to give the title compound (0.23 g, 17% yield). MS (apci) m/z=251.8 (M+H).


Intermediate 157



embedded image


3-amino-5-methyl-2-phenyl-4,5-dihydropyrrolo [3,4-c]pyrazol-6(2H)-one
Step A: Preparation of ethyl 5-amino-4-((methylamino)methyl)-1-phenyl-1H-pyrazole-3-carboxylate

To a stirred solution of ethyl 5-amino-4-formyl-1-phenyl-1H-pyrazole-3-carboxylate (Prepared according to the procedure described in J. Heterocyclic Chemistry, 2010, 47, p. 287-291, 142 mg, 0.548 mmol) in DCM (3 mL) was added 2.0 M MeNH2 in THF (0.822 mL, 1.64 mmol). Two drops of acetic acid was added. The reaction mixture was stirred at ambient temperature overnight. MeOH (0.4 mL) was added followed by NaBH4 (31 mg, 0.82 mmol) portionwise. The reaction was quenched by the slow addition of water. The mixture was extracted with DCM. The combined organic layers were washed with brine, dried and concentrated. The crude was used in the next step without further purification. MS (apci) m/z=275.0 (M+H).


Step B: Preparation of 3-amino-5-methyl-2-phenyl-4,5-dihydropyrrolo[3,4-c]pyrazol-6(2H)-one

To a stirred solution of ethyl 5-amino-4-((methylamino)methyl)-1-phenyl-1H-pyrazole-3-carboxylate (crude, 65 mg, 0.24 mmol) in MeOH (0.5 mL) and THF (0.5 mL) was added 2 N NaOH (0.24 mL, 0.47 mmol). The reaction mixture was stirred at ambient temperature for 4 hours and then concentrated in vacuo. To the residue was added water. The pH was adjusted to 4-5 using 1 N HCl. Water was evaporated under reduced pressure. The crude acid (58 mg) was dissolved in DMF (3 mL). Et3N (66 μL, 0.47 mmol) was added followed by EDCI (90 mg, 0.47 mmol) and HOBt (32 mg, 0.24 mmol). The reaction mixture was stirred at ambient temperature overnight and then partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc. The combined organic layers were washed with water and brine, dried and concentrated. The residue was purified by flash chromatography on silica gel (2% MeOH in DCM) to give the title compound (15 mg, 28%) as a white solid. MS (apci) m/z=228.9 (M+H).


Intermediate 158



embedded image


3-methyl-4-(methylthio)-1-phenyl-1H-pyrazol-5-amine

Prepared according to the method described for Intermediate P109, replacing methyl 2-methoxyacetate with ethyl acetate and replacing acetonitrile with 2-(methylthio)acetonitrile in Step A to afford the product as a brown oil. MS (apci) m/z=220.1 (M+H).


Intermediate 159



embedded image


2-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-2,2-difluoroethanol

Prepared according to the method described for Intermediate P111, replacing acetonitrile with propionitrile and replacing methyl 3-hydroxy-2,2-dimethylpropanoate with ethyl 2,2-difluoro-3-hydroxypropanoate to afford the product as a pale yellow solid. MS (apci) m/z=254.1 (M+H).


Intermediate 160



embedded image


2-(5-amino-1-phenyl-1H-pyrazol-3-yl)-2,2-difluoroethanol

Prepared according to the method described for Intermediate P111, replacing methyl 3-hydroxy-2,2-dimethylpropanoate with ethyl 2,2-difluoro-3-hydroxypropanoate to afford the product as a pale yellow solid. MS (apci) m/z=240.0 (M+H).


Intermediate 161



embedded image


2-(5-amino-1-phenyl-1H-pyrazol-3-yl)ethanol

Prepared according to the method described in Intermediate P111, replacing methyl 3-hydroxy-2,2-dimethylpropanoate with methyl 3-hydroxypropanoate in Step A. MS (apci) m/z=204.1 (M+H).


Intermediate 162



embedded image


1-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-2-methylpropan-2-ol
Step A: Preparation of ethyl 3-hydroxy-3-methylbutanoate

To a solution of lithium bis(trimethylsilyl)amide (1M in THF) (100 mL, 100 mmol) in THF (100 mL) under N2 and cooled to −78° C. was added ethyl acetate (9.74 mL, 100 mmol). The reaction mixture was stirred for 30 minutes, and then acetone (8.81 mL, 120 mmol) was added. The reaction mixture was stirred for 10 minutes, and then quenched with HCl (2M aqueous, 70 mL, 140 mmol) and allowed to warm to ambient temperature. The reaction mixture was extracted with EtOAc (2×150 mL). The organic phases were combined and washed with saturated aqueous NaHCO3 (2×50 mL), dried (MgSO4), filtered and concentrated to afford the product as a yellow oil (12.8 g, 88% yield). 1H NMR (CDCl3) δ 4.18 (q, 3H), 2.49 (s, 2H), 1.29 (m, 9H).


Step B: Preparation of 5-hydroxy-5-methyl-3-oxohexanenitrile

To a solution of propionitrile (1.77 mL, 30.5 mmol) in THF (100 mL) under N2 at −78° C. was added lithium bis(trimethylsilyl)amide (1M in THF) (27.9 mL, 27.9 mmol). Stirred 1 hour, then ethyl 3-hydroxy-3-methylbutanoate (1.86 g, 12.7 mmol) was added. The reaction mixture was stirred at −78° C. for 1 hour, then stirred at 0° C. for 1.5 hours, then diluted with H2O (100 mL) and extracted with Et2O (50 mL). The phases were separated and the basic aqueous phase was neutralized with HCl (6M aqueous, 4.5 mL), then extracted with Et2O (3×75 mL). The combined organic phases were washed with brine (75 mL), dried (MgSO4), filtered, and concentrated to afford the product as a pale yellow oil (1.24 g, 63% yield). 1H NMR (CDCl3) δ 3.54 (m, 1H), 2.89 (s, 2H), 1.50 (d, 3H), 1.32 (s, 3H), 1.31 (s, 3H).


Step C: Preparation of 1-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl)-2-methylpropan-2-ol

To a suspension of phenylhydrazine (0.793 mL, 7.99 mmol) and HCl (5-6M in iPrOH, 1.60 mL, 7.99 mmol) in EtOH (25 mL) was added a solution of 5-hydroxy-2,5-dimethyl-3-oxohexanenitrile (1.24 g, 7.99 mmol) in EtOH (25 mL). The reaction mixture was refluxed for 17 hours, then cooled to ambient temperature, diluted with saturated aqueous NaHCO3 (10 mL), extracted 10:90 MeOH/DCM (3×25 mL), and the combined organic phases were dried (MgSO4), filtered and concentrated. Purified by silica column chromatography eluting with 0-75% acetone/hexanes to afford the title compound as an orange oil (1.13 g, 58% yield). MS (apci) m/z=246.1 (M+H).


The following pyrazole intermediates were prepared according to the method used for the preparation of Intermediate 162, Steps B and C, using the appropriate starting material. For the preparation of Intermediates 168 and 169, the starting material (purchased from Oakwood) was a mixture of cis and trans diastereomers.















Intermediate





#
Structure
Name
MS (apci) m/z







163


embedded image


1-(5-amino-1-phenyl-1H- pyrazol-3-yl)-2- methylpropan-2-ol
232.1 (M+H)





164


embedded image


(S)-1-(5-amino-4-methyl- 1-phenyl-1H-pyrazol-3- yl)propan-2-ol
232.1 (M+H)





165


embedded image


(S)-1-(5-amino-1-phenyl- 1H-pyrazol-3-yl)propan-2- ol
218.1 (M+H)





166


embedded image


(R)-1-(5-amino-4-methyl- 1-phenyl-1H-pyrazol-3- yl)propan-2-ol
232.1 (M+H)





167


embedded image


(R)-1-(5-amino-1-phenyl- 1H-pyrazol-3-yl)propan-2- ol
218.1 (M+H)





168


embedded image


3-(5-amino-4-methyl-1- phenyl-1H-pyrazol-3- yl)cyclobutanol
244.1 (M+H)





169


embedded image


3-(5-amino-1-phenyl-1H- pyrazol-3-yl)cyclobutanol
230.1 (M+H)









Intermediate 170



embedded image


ethyl 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylate

Prepared according to the method described for Intermediate P109, replacing methyl 2-methoxyacetate with diethyl oxalate and replacing acetonitrile with propionitrile in Step A to afford the product as a yellow solid. MS (apci) m/z=246.1 (M+H).


Intermediate 171



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylic acid

To a solution of ethyl 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylate (Intermediate 170, 1.52 mg, 6.21 mmol) in THF (12 mL) and MeOH (6 mL) was added LiOH (2M aq, 9.31 mL, 18.6 mmol). The reaction mixture was stirred at ambient temperature for 19 hours, then partially concentrated under reduced pressure, then neutralized with 6M HCl (3.2 mL), extracted with 10:90 MeOH/DCM (3×25 mL), and the combined organic extracts were washed with brine (50 mL), dried (MgSO4), filtered and concentrated to give the title compound as a yellow solid (1.3 g, 96% yield) MS (apci) m/z=218.1 (M+H).


Intermediate 172



embedded image


5-amino-N,4-dimethyl-1-phenyl-1H-pyrazole-3-carboxamide

To a solution of 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylic acid (Intermediate 171, 223 mg, 1.02 mmol) in acetonitrile (10 mL) were added DIEA (0.71 mL, 4.10 mmol), methanamine hydrochloride (138 mg, 2.05 mmol), DMF (2 mL), and then HATU (428 mg, 1.13 mmol). The reaction mixture was stirred at ambient temperature for 19 hours and then partially concentrated under reduced pressure. The mixture was purified by reverse-phase column chromatography, eluting with 5-60% acetonitrile/water to afford the title compound as a pale yellow solid (182 mg, 77% yield). MS (apci) m/z=231.1 (M+H).


Intermediate 173



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxamide

A solution of 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbonitrile (150 mg, 0.757 mmol) in concentrated H2SO4 (0.5 mL) was stirred at ambient temperature for 17 hours. The reaction mixture was cooled and neutralized by the addition of aqueous NaOH (2M, 11 mL), then extracted 10% MeOH/DCM (5×10 mL), and the combined organic extracts were washed with brine, dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound as a white solid (151 mg, 95% yield). MS (apci) m/z=239.1 (M+Na).


Intermediate 174



embedded image


ethyl 5-amino-3-ethoxy-1-phenyl-1H-pyrazole-4-carboxylate
Step A: Preparation of diethyl 2-cyanomalonate

To a suspension of NaH (60 wt % in mineral oil, 499 mg, 12.49 mmol) in THF (100 mL) under N2 at 0° C. was added diethyl malonate (1.90 mL, 12.49 mmol). The ice bath was removed and the reaction mixture was stirred at ambient temperature for 30 minutes, then cooled to 0° C. and cyanic bromide (5M in MeCN, 2.5 mL, 12.49 mmol) was added. The reaction mixture was stirred at ambient temperature for 19 hours, then diluted with H2O (50 mL), extracted with Et2O (50 mL). The aqueous phase was neutralized with HCl (2M aq, 3 mL) then extracted with DCM (2×50 mL). The combined DCM extracts were dried (MgSO4), filtered, and concentrated to afford the product as a yellow oil (837 mg, 36% yield). 1H NMR (CDCl3) δ 4.46 (s, 1H), 4.35 (q, 4H), 1.35 (t, 6H).


Step B: Preparation of ethyl 5-amino-3-ethoxy-1-phenyl-1H-pyrazole-4-carboxylate

Prepared according to the method described for Intermediate P135, replacing ethyl 2-cyanopropanoate with diethyl 2-cyanomalonate in Step A to afford the product as a brown syrup (400 mg, 32% yield). MS (apci) m/z=276.1 (M+H).


Intermediate 175



embedded image


4-methyl-3-(5-methyl-1,3,4-oxadiazol-2-yl)-1-phenyl-1H-pyrazol-5-amine
Step A: Preparation of N′-acetyl-5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbohydrazide

To a solution of 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylic acid (Intermediate 171, 93 mg, 0.428 mmol) in DCM (5 mL) and DIEA (0.149 mL, 0.856 mmol) was added isobutyl carbonochloridate (0.061 mL, 0.471 mmol). The reaction mixture was stirred at ambient temperature for 1 hour, then acetohydrazide (48 mg, 0.642 mmol) was added. The reaction mixture was stirred at ambient temperature for 18 hours, then diluted with H2O (10 mL), extracted DCM (2×10 mL), dried (MgSO4), filtered and concentrated under reduced pressure to afford the product as a pale yellow solid (119 mg, 101% yield). MS (apci) m/z=274.1 (M+H).


Step B: Preparation of 4-methyl-3-(5-methyl-1,3,4-oxadiazol-2-yl)-1-phenyl-1H-pyrazol-5-amine

A mixture of N′-acetyl-5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbohydrazide (117 mg, 0.428 mmol) and POCl3 (0.5 mL) was heated in a pressure tube to 90° C. for 1 hour. The reaction mixture was transferred to a separatory funnel with EtOAc (5 mL), then diluted with saturated aqueous NaHCO3 (20 mL), extracted with EtOAc (2×15 mL), dried (MgSO4), filtered and concentrated. The residue was purified by silica column chromatography eluting with 0-75% acetone/hexanes to afford the title compound as a yellow solid (19.6 mg, 18% yield). MS (apci) m/z=256.1 (M+H).


Intermediate 176



embedded image


4-methyl-3-(3-methyl-1,2,4-oxadiazol-5-yl)-1-phenyl-1H-pyrazol-5-amine

To a suspension of NaH (60% in mineral oil, 36 mg, 0.897 mmol) in THF (5 mL) under N2 was added N-hydroxyacetimidamide (66 mg, 0.897 mmol). The reaction mixture was heated to reflux for 1 hour, then cooled to ambient temperature and ethyl 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylate (Intermediate 170, 200 mg, 0.815 mmol) was added. The reaction mixture was heated to reflux for 18 hours, then cooled to ambient temperature and additional NaH (60% in mineral oil, 18 mg, 0.449 mmol) was added. The reaction mixture was heated to reflux for 4 hours, then diluted with H2O (10 mL), extracted DCM (2×15 mL), and the combined organic extracts were dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography eluting with 0-50% acetone/hexanes to afford the title compound as an orange solid (84 mg, 40% yield). MS (apci) m/z=256.1 (M+H).


Intermediate 177



embedded image


3-(3-methyl-1,2,4-oxadiazol-5-yl)-1-phenyl-1H-pyrazol-5-amine

Prepared according to the method described in Intermediate 176, replacing ethyl 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylate with ethyl 5-amino-1-phenyl-1H-pyrazole-3-carboxylate (Nanjing Chemlin Chemical Co.) to afford the product as a tan solid (83 mg, 53% yield). MS (apci) m/z=242.1 (M+H).


Intermediate 178



embedded image


4-methyl-1-phenyl-3-(3-(trifluoromethyl)-1,2,4-oxadiazol-5-yl)-1H-pyrazol-5-amine
Step A: Preparation of 2,2,2-trifluoro-N′-hydroxyacetimidamide

To a suspension of hydroxylamine hydrochloride (5.45 g, 78.4 mmol) in MeOH (100 mL) was added NaOMe (25 wt % solution in MeOH, 17.9 mL, 78.4 mmol) and the mixture stirred at ambient temperature for 10 minutes, then filtered and the solid was washed with MeOH. The filtrate was cooled to 0° C. and then 2,2,2-trifluoroacetonitrile (7.45 g, 78.4 mmol) gas was bubbled into the solution over 30 minutes. The reaction mixture was then allowed to warm to ambient temperature for 19 hours. The solution was concentrated under reduced pressure to 50 mL and the solids were filtered. The filtrate was concentrated, re-suspended in cold MeOH, and filtered. The filtrate was concentrated, again re-suspended in cold MeOH, and filtered. The filtrate was concentrated to give the product as a waxy white solid (6.7 g, 67% yield). 1H NMR (CD3CN) δ 8.32 (s, 1H), 5.25 (br s, 2H). 19F NMR (CD3CN) δ-71.8 (s).


Step B: Preparation of 4-methyl-1-phenyl-3-(3-(trifluoromethyl)-1,2,4-oxadiazol-5-yl)-1H-pyrazol-5-amine

To a suspension of NaH (60% in mineral oil, 356 mg, 0.897 mmol) in THF (5 mL, 0.815 mmol) under N2 was added 2,2,2-trifluoro-N′-hydroxyacetimidamide (115 mg, 0.897 mmol). The reaction mixture was heated to reflux for 1 hour, then cooled to ambient temperature and powdered 4 A molecular sieves (200 mg) and ethyl 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxylate (Intermediate 170; 200 mg, 0.815 mmol) were added and heated to reflux. The reaction mixture was heated to reflux for 18 hours, then filtered, diluted with H2O (15 mL), extracted DCM (2×25 mL), and the combined organic extracts were washed with brine (25 mL), dried (MgSO4), filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography eluting with 0-50% acetone/hexanes to afford the title compound as a white solid (44 mg, 17% yield). MS (apci) m/z=310.1 (M+H).


Intermediate 179



embedded image


2-phenyl-2H-indazol-3-amine
Step A: Preparation of 1-(2-iodophenyl)-2-phenyldiazene

To a solution of 2-iodoaniline (1.00 g, 4.57 mmol) in acetic acid (46 mL) was added nitrosobenzene (0.880 g, 8.22 mmol) and the mixture was heated at 85° C. for 16 hours. The mixture was cooled to ambient temperature, poured into water and slowly treated with saturated NaHCO3 until basic. The mixture was extracted with EtOAc (3×) and the combined extracts were washed with water, saturated NaCl and dried over MgSO4. The solution was filtered, concentrated and the residue purified by reverse phase chromatography to provide the title compound as a red solid (0.880 g, 63% yield). 1H NMR (CDCl3) δ 7.23-7.39 (m, 3H), 7.64 (d, 1H), 7.56-7.51 (m, 3H), 7.45 (t, 1H), 7.1 (t, 1H).


Step B: 2-(phenyldiazenyl)benzonitrile

To a solution of 1-(2-iodophenyl)-2-phenyldiazene (0.44 g, 1.4 mmol) in 1-propanol (14 mL) was added CuCN (0.900 g, 10.0 mmol) and the reaction was heated at reflux for 16 hours. The mixture was cooled to ambient temperature, filtered and the collected solid washed with CH2Cl2. The combined filtrate and washes were concentrated to provide the title compound as red-orange solid that was dried in vacuum (0.280 g, 95% yield). 1H NMR (CDCl3) δ 8.03-8.06 (m, 2H), 7.88 (dd, 2H), 7.71 (t, 1H), 7.54-7.58 (m, 4H).


Step C: 2-phenyl-2H-indazol-3-amine

A mixture of 2-(phenyldiazenyl)benzonitrile (0.28 g, 1.35 mmol) and SnCl2 dihydrate (0.562 mL, 6.76 mmol) in EtOH (14 mL) was heated at reflux for 16 hours. The mixture was cooled to ambient temperature and concentrated. The residue was diluted with EtOAc and water and filtered. The aqueous layer was removed and the EtOAc layer was washed with water. The combined aqueous fractions were basified with saturated NaHCO3 and extracted with CH2Cl2 (2×). The combined organic layers were dried over MgSO4, filtered and concentrated to provide the title compound as a light purple solid that was dried in vacuum (0.241 g, 85% yield). 1H NMR (CDCl3) δ 7.69 (d, 2H), 7.52-7.58 (m, 3H), 7.47 (d, 2H), 7.26 (t, 1H), 6.90 (t, 1H), 4.28 (br s, 2H).


Intermediate 180



embedded image


3-ethoxy-4-methyl-1-(pyrazin-2-yl)-1H-pyrazol-5-amine
Step A: 5-amino-4-methyl-1-(pyrazin-2-yl)-1H-pyrazol-3(2H)-one

To a mixture of 2-hydrazinylpyrazine (0.551 g, 5.00 mmol) and ethyl 2-cyanopropanoate (0.669 g, 5.00 mmol) in abs. EtOH (10 mL) was added 3M NaOEt in EtOH (0.167 mL, 0.501 mmol) and the mixture was heated at reflux for 64 hours. The mixture was concentrated and the residual yellow-brown solid was treated with EtOAc (30 mL) and sonicated. The resulting tan suspension was stirred vigorously for 8 hours. The solid was collected via vacuum filtration, washed with EtOAc and dried in vacuum to afford the title compound as a light tan powder (682 mg, 71%). 1H NMR (DMSO d6) δ 10.3 (br s, 1H), 8.82 (s, 1H), 8.30 (d, 2H), 6.55 (s, 2H), 1.71 (s, 3H).


Step B: 3-ethoxy-4-methyl-1-(pyrazin-2-yl)-1H-pyrazol-5-amine

A mixture of 5-amino-4-methyl-1-(pyrazin-2-yl)-1H-pyrazol-3(2H)-one (382 mg, 2.00 mmol) and powdered K2CO3 (552 mg, 4.00 mmol) in dry DMF (3.0 mL) was stirred at ambient temperature for 10 minutes. The mixture was cooled to 0° C. and bromoethane (229 mg, 2.10 mmol) was added. The mixture was allowed to reach ambient temperature and was stirred 24 hours. The reaction mixture poured into cold H2O (12 mL), allowed to reach ambient temperature and was extracted with EtOAc (3×). The combined extracts were washed with saturated NaCl (2×), dried over MgSO4 and activated carbon. The dried solution was diluted with and equal volume of hexanes and filtered through a SiO2 plug capped with a MgSO4 layer eluting with 50% EtOAc-hexanes. The filtrate was concentrated and the residual yellow solid was washed with hexanes (3×) and dried in vacuum to afford the title compound as a light yellow crystalline solid (195 mg, 45%). 1H NMR (CDCl3) δ 9.10 (s, 1H), 8.23 (s, 1H), 8.14 (s, 1H), 5.50 (br s, 2H), 4.33 (q, 2H), 1.80 (s, 3H), 1.42 (t, 3H).


Intermediate 181



embedded image


2-(pyridazin-4-yl)-2,4,5,6-tetrahydrocyclopenta[c]pyrazol-3-amine

A suspension of 4-hydrazinylpyridazine hydrobromide (0.368 g, 1.93 mmol) in absolute EtOH (5 mL) was treated with 2-oxocyclopentanecarbonitrile (0.191 g, 1.75 mmol) and the mixture was heated at reflux for 22 hours. The mixture was cooled to ambient temperature and was concentrated to an orange solid. The solid was suspended in 1M NaOH and stirred for 10 minutes. The solid was collected, washed thoroughly with H2O and Et2O and dried in vacuum to furnish title compound as a tan powder (0.323 g, 92%). MS (apci) m/z=202.1 (M+H).


Intermediate 182



embedded image


(5-amino-1-phenyl-1H-pyrazol-3-yl)methanol
Step A: Ethyl 2-(tert-butyldimethylsilyloxy)acetate

A mixture of ethyl 2-hydroxyacetate (3.00 g, 28.8 mmol), TBDMS-Cl (5.21 g, 34.6 mmol) and imidazole (2.55 g, 37.5 mmol) was stirred at ambient temperature for 60 hours. The mixture was concentrated and the residue was purified by SiO2 chromatography eluting with 10% EtOAc-hexanes to provide the title compound as a colorless oil (4.12 g, 65%). 1H NMR (CDCl3) δ 4.12 (s, 2H), 4.09 (q, 2H), 1.17 (t, 3H), 0.18 (s, 9H), 0.00 (s, 6H).


Step B: (5-amino-1-phenyl-1H-pyrazol-3-yl)methanol

A solution of acetonitrile (0.526 mL, 10.1 mmol) in dry THF (20.4 mL, 9.16 mmol) was cooled to −78° C. and 2.5M nBuLi in hexanes (4.21 mL, 10.5 mmol) was added dropwise. The reaction mixture was stirred for 15 minutes and ethyl 2-(tert-butyldimethylsilyloxy)acetate (2.00 g, 9.16 mmol) was added. The reaction mixture was allowed to warm to ambient temperature and was stirred for 2 hours. The reaction mixture was diluted with ice water and was concentrated. The residual aqueous mixture was acidified to pH=5 and extracted with EtOAc (3×). The combined organics were washed with brine, dried over MgSO4, filtered and concentrated. The residual brown oil was dissolved in MeOH (23 mL) and phenyl hydrazine (0.907 mL, 9.14 mmol) was added. The mixture was treated with concentrated HCl (3.81 mL, 45.7 mmol) and heated at reflux for 18 hours. Upon cooling, the mixture was concentrated and the residue was partitioned into in H2O and CH2Cl2. The mixture was filtered and the organic layer was removed from the filtrate. The aqueous portion was washed with CH2Cl2 and was treated with saturated NaHCO3 until basic. The aqueous mixture was extracted with CH2Cl2 (3×) and the combined organic fractions were dried over MgSO4, filtered and concentrated. The residue was purified by silica column chromatography using 70-100% EtOAc/hexanes gradient elution followed by 0-5% MeOH/EtOAc. The product pools were combined and concentrated to give the title compound as a yellow foam (0.760 g, 44% yield). MS (apci) m/z=190.1 (M+H).


Intermediate 183



embedded image


4-methyl-3-((1-methyl-1H-1,2,4-triazol-3-yl)methoxy)-1-phenyl-1H-pyrazol-5-amine

The title compound was prepared by the method as described for Intermediate P135, substituting bromoethane with 3-(chloromethyl)-1-methyl-1H-1,2,4-triazole hydrochloride. The product was isolated as a gold syrup (110 mg, 27%). MS (apci) m/z=285.1 (M+H).


Intermediate 184



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl dimethylcarbamate

A mixture of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3 (2H)-one (Intermediate P135 Step A, 0.378 g, 2.00 mmol) and powdered K2CO3 (0.553 g, 4.00 mmol) in dry DMF (4 mL) was stirred at ambient temperature for 5 minutes. Dimethylcarbamoyl chloride (0.206 mL, 2.20 mmol) was added and the mixture was stirred for 6 hours. The mixture was poured into chilled H2O (40 mL) and was extracted with EtOAc (3×). The combined extracts were washed with saturated NaCl (2×), dried over MgSO4 and filtered through a SiO2 plug capped with a MgSO4 layer (EtOAc elution). The filtrate was concentrated and the residue dried in vacuum to give the title compound as a light gold syrup (0.507 g, 97%). MS (apci) m/z=261.1 (M+H).


Intermediate 185



embedded image


5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl morpholine-4-carboxylate

The title compound was prepared using morpholine-4-carbonyl chloride in the procedure outlined for 5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yl dimethylcarbamate (Intermediate 184). The compound was isolated as a light yellow wax (0.285 g, 47%). 1H NMR (CDCl3) δ 7.54 (d, 2H), 7.43 (t, 2H), 7.31 (t, 1H), 3.66-3.78 (m, 8H), 3.57 (br s, 2H), 1.85 (s, 3H).


Intermediate 186



embedded image


(S)-3-(2-((tert-butyldimethylsilyl)oxy)-3-methoxypropoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine
Step A: (S)-1-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yloxy)-3-methoxypropan-2-ol

A mixture of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (P135 Step A, 1.21 g, 6.40 mmol) and powdered K2CO3 (1.77 g, 12.8 mmol) in dry DMF (12 mL) was stirred at ambient temperature for 10 minutes. (S)-2-(methoxymethyl)oxirane (0.622 mL, 6.72 mmol) was added and the mixture was stirred at 80° C. for 6 hours. The mixture was cooled to ambient temperature, poured into chilled H2O (25 mL) and extracted with EtOAc (3×). The combined extracts were washed with saturated NaCl (2×), dried over MgSO4 and filtered through a SiO2 plug capped with a layer of MgSO4 eluting with EtOAc. The filtrate was concentrated to give the title compound as a colorless, viscous oil (701 mg, 40%). MS (apci) m/z=278.1 (M+H).


Step B: (S)-3-(2-((tert-butyldimethylsilyl)oxy)-3-methoxypropoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine

To a solution of TBDMS-Cl (725 mg, 4.81 mmol) and imidazole (390 mg, 5.72 mmol) in dry DMF (7.0 mL) was added (S)-1-(5-amino-4-methyl-1-phenyl-1H-pyrazol-3-yloxy)-3-methoxypropan-2-ol (635 mg, 2.29 mmol) in dry DMF (2 mL). The mixture stirred at ambient temperature for 2.5 hours. The mixture added to H2O (70 mL), mixed for 5 minutes and extracted with Et2O (3×). The combined extracts were washed with saturated NaCl (2×) and dried over MgSO4. The dried solution was filtered through a SiO2 plug capped with a layer of MgSO4 (Et2O elution). The filtrate was concentrated to give the title compound as a colorless oil that was dried in vacuum (940 mg, 105%). MS (apci) m/z=392.2 (M+H). 1H NMR (CDCl3) δ 7.50 (d, 2H), 7.40 (t, 2H), 7.23 (t, 1H), 4.09-4.30 (m, 3H), 3.57 (br s, 2H), 3.38-3.44 (m, 2H), 3.32 (s, 3H), 1.83 (s, 3H), 0.88 (s, 9H), 0.11 (s, 6H).


Intermediate 187



embedded image


(R)-3-(2-((tert-butyldimethylsilyl)oxy)-3-methoxypropoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine

The title compound was prepared using the procedure described for (S)-3-(2-((tert-butyldimethylsilyl)oxy)-3-methoxypropoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine (Intermediate 186) substituting (S)-2-(methoxymethyl)oxirane with (R)-2-(methoxymethyl)oxirane in Step A. The product was obtained as a colorless syrup (921 mg, 38% over 2 steps). MS (apci) m/z=392.2 (M+H). 1H NMR (CDCl3) δ 7.50 (d, 2H), 7.40 (t, 2H), 7.23 (t, 1H), 4.09-4.30 (m, 3H), 3.57 (br s, 2H), 3.38-3.44 (m, 2H), 3.32 (s, 3H), 1.83 (s, 3H), 0.88 (s, 9H), 0.11 (s, 6H).


Intermediate 188



embedded image


tert-butyl 4-((5-amino-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate
Step A: tert-butyl 4-(2-ethoxy-2-oxoethoxy)piperidine-1-carboxylate

A solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (2.00 g, 9.94 mmol) in dry THF (25 mL) was cooled to 0° C. and KOtBu (1.12 g, 9.94 mmol) was added. The mixture was allowed to reach ambient temperature and was stirred for 10 minutes. The mixture was cooled to 0° C. and ethyl 2-bromoacetate (1.65 mL, 14.9 mmol) was added dropwise. The reaction was allowed to reach ambient temperature and was stirred for 17 hours. The mixture was partitioned into in H2O and EtOAc, mixed and the organic layer was removed. The organic layer was dried over MgSO4, filtered and concentrated. The residual thick yellow oil was purified by silica chromatography using a 10-25% EtOAc/hexanes gradient elution to afford the title compound as a colorless oil (0.967 g, 34% yield). 1H NMR (CDCl3) δ 4.22 (q, 2H), 4.12 (s, 2H), 3.67-3.84 (m, 2H), 3.52-3.63 (m, 1H), 3.05-3.11 (m, 2H), 1.81-1.90 (m, 2H), 1.53-1.62 (m, 2H), 1.45 (s, 9H), 1.29 (t, 3H).


Step B: tert-butyl 4-((5-amino-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate

A solution of diisopropylamine (1.08 mL, 7.74 mmol) in dry THF (5 mL) was cooled to 0° C. and 2.5M nBuLi in hexanes (2.96 mL, 7.41 mmol) was slowly added. The mixture was stirred at 0° C. for 10 minutes and was cooled to −78° C. Acetonitrile (0.404 mL, 7.74 mmol) was added and the mixture was stirred for 15 minutes. A solution of tert-butyl 4-(2-ethoxy-2-oxoethoxy)piperidine-1-carboxylate (0.967 g, 3.37 mmol) in THF (2.5 mL) was added and the mixture was stirred at −78° C. for 1 hour. The mixture was allowed to reach ambient temperature, was quenched with ice water and concentrated. The residual aqueous mixture was neutralized with 2M HCl and was extracted with CH2Cl2 (3×). The combined organic fractions were dried over MgSO4, filtered and concentrated to provide the crude cyano-ketone as a yellow oil that was used immediately in the next step.


Step C: tert-butyl 4-((5-amino-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate

The crude oil obtained in Step B was dissolved in EtOH (17 mL) and phenylhydrazine (0.396 mL, 3.99 mmol) was added. The mixture was heated at 60° C. for 60 hours, was cooled to ambient temperature and was concentrated. The residue was partitioned into EtOAc and water, mixed and the organic layer removed. The aqueous layer was extracted with EtOAc (2×) and the combined EtOAc portions were dried over MgSO4, filtered and concentrated. The residual orange oil was purified by silica chromatography using a 10-100% EtOAc/hexanes gradient elution. The pooled product fractions were concentrated and the residual yellow-orange oil was re-purified by reverse phase HPLC using a 0-100% acetonitrile/water gradient to provide the title compound as an orange foam (0.264 g, 21% yield). MS (apci) m/z=373.2 (M+H).


Intermediate 189



embedded image


1-phenyl-3-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-5-amine
Step A: 3-oxo-3-(tetrahydro-2H-pyran-4-yl)propanenitrile

A 1M solution of LHMDS in dry THF (26.3 mL, 26.3 mmol) was cooled to −78° C. and acetonitrile (1.43 mL, 27.5 mmol) was added dropwise over 2 minutes. The mixture was stirred at −78° C. for 1 hour and a solution of methyl tetrahydro-2H-pyran-4-carboxylate (3.41 mL, 25.0 mmol) in dry THF (12 mL) was added. The mixture was stirred for 1 hour, the dry ice bath was removed and the mixture allowed to reach ambient temperature. The mixture was poured into chilled H2O (250 mL) and was extracted with Et2O (3×). The aqueous portion was cooled to 0° C. and 6M HCl was added dropwise to pH=3 (starting pH=12). The mixture was extracted with EtOAc (3×) and the combined extracts were dried over MgSO4. The solution eluted through a SiO2 plug eluting with EtOAc. The filtrate was concentrated to give the title compound as a colorless oil (2.52 g, 66%). 1H NMR (CDCl3) δ 3.99-4.06 (m, 2H), 3.54 (s, 2H), 3.46 (t, 2H), 2.76-2.86 (m, 1H), 1.70-1.86 (m, 4H).


Step B: 1-phenyl-3-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-5-amine

To a solution of 3-oxo-3-(tetrahydro-2H-pyran-4-yl)propanenitrile (2.30 g, 12.8 mmol) in absolute EtOH (35 mL) was added phenylhydrazine hydrochloride (2.21 g, 15.3 mmol) and the mixture was heated at reflux until complete by TLC (5 hours). The mixture was cooled to ambient temperature and was concentrated. The residue was partitioned in H2O (75 mL) and EtOAc (40 mL). 2M NaOH was added to pH=5 with vigorous mixing, the organic layer was removed and the aqueous was extracted with EtOAc (2×). The combined EtOAc fractions were washed with H2O and saturated NaCl. The solution was diluted with an equal volume of hexanes, dried over MgSO4/activated carbon and eluted through a SiO2 plug eluting with 50% EtOAc-hexanes. The filtrate was concentrated to give a gold syrup. The syrup was treated with Et2O and stirred until a fine, granular suspension formed. The solid was collected, washed with Et2O and dried in vacuum to furnish the title compound as a white solid (2.01 g, 65%). 1H NMR (CDCl3) δ 7.55 (d, 2H), 7.46 (t, 2H), 7.32 (t, 1H), 5.49 (s, 1H), 4.00-4.08 (m, 2H), 3.97 (br s, 2H), 3.52 (dt, 2H), 2.86 (m, 1H) 1.73-1.93 (m, 4H).


The following compounds were prepared according to the method used for the preparation of 1-phenyl-3-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-5-amine (Intermediate 189) using either acetonitrile or propiononitrile in Step A in conjunction with the appropriate ester.














Intermediate #
Structure
Data







190


embedded image


MS (apci) m/z = 343.1 (M + H)





191


embedded image


MS (apci) m/z = 258.0 (M + H)





192


embedded image



1H NMR (CDCl3) δ 7.62 (d, 2H), 7.50 (t, 2H), 7.37 (t, 1H), 5.72 (s, 1H), 3.91 (br s, 2H), 2.58 (s, 3H), 2.44 (s, 3H).






193


embedded image



1H NMR (CDCl3) δ 7.60 (d, 2H), 7.49 (t, 2H), 7.37 (t, 1H), 6.45 (s, 1H), 3.67 (br s, 2H), 2.45 (s, 3H), 2.24 (s, 3H).






194


embedded image



1H NMR (CDCl3) δ 7.45-7.56 (m, 4H), 7.35 (t, 1H), 4.00-4.06 (m, 2H), 3.88 (dt, 2H), 3.62 (br s, 2H), 2.18-2.34 (m, 4H), 2.11 (s, 3H).






195


embedded image


MS (apci) m/z = 343.2 (M + H)





196


embedded image


MS (apci) m/z = 343.2 (M + H)





197


embedded image


MS (apci) m/z = 329.2 (M + H)





198


embedded image


MS (apci) m/z = 329.2 (M + H)









Intermediate 199



embedded image


Phenyl 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-ylcarbamate
Step A: ethyl 1-methyl-1H-pyrazole-4-carboxylate

To a 3000-mL three-necked flask was added ethyl 2-formyl-3-oxopropanoate (100 g, 694 mmol), followed by anhydrous 200-proof EtOH (694 mL) to obtain a clear yellowish solution. The reaction was cooled in an ice bath to 5° C., and then methylhydrazine (35.8 mL, 680 mmol) was added dropwise. A vigorous exotherm was observed during hydrazine addition and the temperature was kept below 12° C. by controlling the addition rate. After the hydrazine addition was complete, the ice bath was removed, and the reaction was allowed to stir at ambient temperature overnight. The reaction was concentrated on a rotary evaporator to a crude orange oil. The crude was taken up in DCM and re-concentrated, then on high vacuum for 2 days to yield tan orange oil. LC/MS and 1H NMR showed essentially pure ethyl 1-methyl-1H-pyrazole-4-carboxylate (106 g, 99.1%).


Step B: 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile

To a four-necked 5-liter round bottomed flask fitted with an overhead stirrer and addition funnel was charged LHMDS (1444 mL, 1444 mmol) (1.0 M in THF). The solution was cooled in an acetone/dry ice bath first (internal temperature of −79° C.) under nitrogen, followed by slow addition of propiononitrile (103 mL, 1444 mmol) via dropping funnel. The mixture was stirred at −80° C. for 90 minutes. A solution of ethyl 1-methyl-1H-pyrazole-4-carboxylate (106 g, 688 mmol) in anhydrous THF (500 mL) was then introduced dropwise via an addition funnel (addition time: about 45 minutes; internal temperature during addition remained below −76° C.). After the addition was complete, the reaction was allowed to slowly warm to ambient temperature and stirred overnight. An orange glass deposited on the bottom of the flask. The organics were decanted and the glass was dissolved in warm water. The mixture was washed with ether (3×1000 mL). The aqueous phase was then pH-adjusted to 5 (pH paper) using concentrated HCl and saturated bicarbonate solution The aqueous layer was extracted with DCM (3×1000 mL). The combined organic extracts were dried over MgSO4 filtered and concentrated to yield the 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile as an amber oil (92 g, 82%). MS (apci) m/z=162.1 (M−H).


Step C: 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine

A 3 L, 3 necked round bottomed flask was charged with 2-methyl-3-(1-methyl-1H-pyrazol-4-yl)-3-oxopropanenitrile (60 g, 368 mmol) absolute anhydrous ethanol (1000 mL) and phenylhydrazine hydrochloride (58 g, 404 mmol) at ambient temperature to form a yellowish suspension. The reaction vessel was equipped with a water condenser and refluxed (using a heating mantle) overnight. The reaction was concentrated and 1M NaOH (1 L) was added and the solid was broken up and collected. The solid was washed with water and hexanes. A second crop crashed out in the filtrate and was collected. The combined solids were crushed and triturated with ether (500 mL). The solid was collected filtration, washed with hexanes and air dried under vacuum to provide 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine (93 g, 100%).


Step D: phenyl 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-ylcarbamate

In a 3 L, round bottomed flask was charged with 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine (50 g, 197.4 mmol) and EtOAc (1000 mL) to obtain a clear brownish solution. To this was added NaOH (2M aq) (500 mL) in one portion to obtain a turbid mixture (both the aqueous and organic layers were clear but a precipitate was observed in between the two layers). After 3 minutes, phenyl carbonochloridate (74.29 mL, 592.2 mmol) was added slowly at ambient temperature exotherm to 33° C. The reaction stirred at ambient temperature for 2 hours. Additional phenyl carbonochloridate (10 mL) was added. After 30 minutes the organics were separated, washed with brine and concentrated in vacuo. The product was purified by silica gel chromatography (eluting with 75% ethyl acetate in hexanes) to provide phenyl 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-ylcarbamate (60 g, 81.4%).


Intermediate 200



embedded image


phenyl 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-ylcarbamate

A 3 L, round bottomed flask was charged with 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-amine (50 g, 197.4 mmol) and EtOAc (1000 mL) to obtain a clear brownish solution. To this was added NaOH (2M aq) (500 mL) in one portion to obtain a turbid mixture (the aqueous and organic layers were clear, but a precipitate was observed in between the two layers). After 3 minutes, phenyl carbonochloridate (74.29 mL, 592.2 mmol) was added slowly at ambient temperature (the temperature of the reaction mixture increased to 33° C. during the addition). The reaction stirred at ambient temperature for 2 hours. Additional phenyl carbonochloridate (10 mL) was added. After 30 minutes the organics layers were separated, washed with brine and concentrated in vacuo. The residue was purified by silica gel chromatography (eluting with 75% ethyl acetate in hexanes) to provide phenyl 1′,4-dimethyl-1-phenyl-1H,1′H-3,4′-bipyrazol-5-ylcarbamate (60 g, 81.4%).


Intermediate 201



embedded image


phenyl (4-chloro-3-ethoxy-1-phenyl-1H-pyrazol-5-yl)carbamate
Step A: Preparation of phenyl (3-ethoxy-1-phenyl-1H-pyrazol-5-yl)carbamate

To a suspension of 3-ethoxy-1-phenyl-1H-pyrazol-5-amine (Intermediate P139, 169 mg, 0.832 mmol) in EtOAc (5 mL) at 0° C. was added 2.0 M aqueous NaOH solution (1.25 mL, 2.50 mmol), followed by dropwise addition of phenyl carbonochloridate (0.178 mL, 1.41 mmol). The reaction was stirred at ambient temperature for 15 hours. The reaction mixture was diluted with EtOAc and phase-separated. The organic layer was washed with water and brine, dried over MgSO4, and concentrated. The residue was purified by flash chromatography on silica gel (6:1 hexanes:EtOAc) to give the title compound (219 mg, 81% yield). MS (apci) m/z=324.1 (M+H).


Step B: Preparation of phenyl (4-chloro-3-ethoxy-1-phenyl-1H-pyrazol-5-yl)carbamate

To a solution of phenyl 3-ethoxy-1-phenyl-1H-pyrazol-5-ylcarbamate (92 mg, 0.28 mmol) and pyridinium 4-methylbenzenesulfonate (7.2 mg, 0.028 mmol) in DCM (2 mL) was added N-chlorosuccinimide (42 mg, 0.31 mmol) at ambient temperature. The mixture was stirred at ambient temperature for 2 days and then concentrated under reduced pressure.


The residue was purified by flash chromatography on silica gel (9:1, hexanes/EtOAc) to give the title compound (76 mg, 75% yield). MS (apci) m/z=358.1 (M+H).


Intermediate 203



embedded image


Phenyl (4-bromo-3-(2-hydroxy-2-methylpropoxy)-1-phenyl-1H-pyrazol-5-yl)carbamate
Step A: Preparation of 5-amino-1-phenyl-1H-pyrazol-3(2H)-one

Prepared according to the method described for Intermediate P1, replacing 4,4-dimethyl-3-oxopentanenitrile with ethyl 2-cyanoacetate, and substituting phenylhydrazine for ethyl 3-hydrazinylbenzoate hydrochloride. MS (apci) m/z=176.0 (M+H).


Step B: Preparation of 1-((5-amino-1-phenyl-1H-pyrazol-3-yl)oxy)-2-methylpropan-2-ol

A mixture of 5-amino-1-phenyl-1H-pyrazol-3(2H)-one (0.330 g, 1.88 mmol), 2,2-dimethyloxirane (0.143 g, 1.98 mmol) and K2CO3 (0.521 g, 3.77 mmol) in DMA (5 mL) was heated at 80° C. for 3 days. After cooling, the reaction mixture was diluted with EtOAc, washed with water and brine and dried over MgSO4. The mixture was filtered through a pad of SiO2 eluting with EtOAc to yield the title compound. MS (apci) m/z=248.1 (M+H).


Step C: Preparation of phenyl (3-(2-hydroxy-2-methylpropoxy)-1-phenyl-1H-pyrazol-5-yl)carbamate

Prepared according to the method described for Intermediate 201. Step A using 1-((5-amino-1-phenyl-1H-pyrazol-3-yl)oxy)-2-methylpropan-2-ol as a replacement for 3-ethoxy-1-phenyl-1H-pyrazol-5-amine. MS (apci) m/z=368.1 (M+H).


Step D: Preparation of phenyl (4-bromo-3-(2-hydroxy-2-methylpropoxy)-1-phenyl-1H-pyrazol-5-yl)carbamate

Prepared according to the method described for Intermediate 201, Step B using N-bromosuccinimide as a replacement for N-chlorosuccinimide, and substituting phenyl (3-(2-hydroxy-2-methylpropoxy)-1-phenyl-1H-pyrazol-5-yl)carbamate for phenyl 3-ethoxy-1-phenyl-1H-pyrazol-5-ylcarbamate. MS (apci) m/z=446.1 (M+H).


The following compounds prepared according to the method describe for the preparation of Intermediate 200, using the appropriate amino pyrazole intermediate:















Intermediate





#
Structure
Name
Data







204


embedded image


phenyl 3-(3- methoxypropyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 366.1 (M + H).





205


embedded image


phenyl 3-(1,1-difluoro- 2-hydroxyethyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 374.1 (M + H).





206


embedded image


(S)-phenyl 3-(2- hydroxypropyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 352.1 (M + H).





207


embedded image


(R)-phenyl 3-(2- hydroxypropyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 352.1 (M + H).





208


embedded image


phenyl 3-(2-hydroxy-2- methylpropyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 366.2 (M + H).





209


embedded image


phenyl 3-(3- hydroxycyclobutyl)-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 364.2 (M + H).





210


embedded image


phenyl 3-(2- hydroxyethyl)-4-methyl- 1-phenyl-1H-pyrazol-5- ylcarbamate
MS (apci) m/z = 338.1 (M + H).





211


embedded image


ethyl 4-methyl-5- (phenoxycarbonylamino)- 1-phenyl-1H-pyrazole- 3-carboxylate
MS (apci) m/z = 366.1 (M + H).





212


embedded image


phenyl 4-methyl-3- (methylcarbamoyl)-1- phenyl-1H-pyrazol-5- yl carbamate
MS (apci) m/z = 351.1 (M + H).





213


embedded image


phenyl 3-carbamoyl-4- methyl-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 337.1 (M + H).





214


embedded image


phenyl (4-methyl-3-(5- methyl-1,3,4-oxadiazol- 2-yl)-1-phenyl-1H- pyrazol-5-yl)carbamate
MS (apci) m/z = 376.1 (M + H).





215


embedded image


phenyl 4-methyl-3-(3- methyl-1,2,4-oxadiazol- 5-yl)-1-phenyl-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 376.1 (M + H).





216


embedded image


phenyl 4-methyl-1- phenyl-3-(3- (trifluoromethyl)-1,2,4- oxadiazol-5-yl)-1H- pyrazol-5-ylcarbamate
MS (apci) m/z = 430.1 (M + H).





217


embedded image


tert-butyl 4-(5- ((phenoxycarbonyl)amino)- 1-phenyl-1H-pyrazol- 3-yl)piperidine-1- carboxylate
MS (apci) m/z = 463.3 (M + H)





218


embedded image


phenyl (4-methyl-1- phenyl-3-(tetrahydro- 2H-pyran-4-yl)-1H- pyrazol-5-yl)carbamate
MS (apci) m/z = 378.2 (M + H)





219


embedded image


phenyl (3-(3,5- dimethylisoxazol-4-yl)- 1-phenyl-1H-pyrazol-5- yl)carbamate

1H NMR (CDCl3) δ 7.56- 7.64 (m, 4H), 7.48-7.52 (m, 1H), 7.40 (t, 2H), 7.26 (t, 2H), 7.16 (br s, 2H), 6.71 (br s, 1H), 2.60 (s, 3H) 2.46 (s, 3H)






220


embedded image


phenyl (4-methyl-3-(5- methylisoxazol-3-yl)-1- phenyl-1H-pyrazol-5- yl)carbamate

1H NMR (CDCl3) δ 7.54 (d. 2H), 7.49 (t, 2H), 7.41 (t, 1H), 7.33 (br s, 2H), 7.20 (br s, 1H), 7.08 (br s, 1H), 6.74 (br s, 1H), 6.66 (br s, 1H), 6.48 (s, 1H), 2.45 (s, 3H) 2.34 (s, 3H)






221


embedded image


phenyl (3-(4- cyanotetrahydro-2H- pyran-4-yl)-4-methyl-1- phenyl-1H-pyrazol-5- yl)carbamate

1H NMR (CDCl3) δ 7.06- 7.56 (m, 9H), 6.75 (br s, 1H), 6.51 (s, 1H), 4.04 (d, 2H) 3.89 (t, 2H), 2.20- 2.39 (m, 4H), 2.28 (s, 3H)






222


embedded image


(R)-tert-butyl 2-(4- methyl-5-((phenoxy- carbonyl)amino)-1- phenyl-1H-pyrazol-3- yl)pyrrolidine-1- carboxylate
MS (apci) m/z = 463.2 (M + H)





223


embedded image


(S)-tert-butyl 2-(4- methyl-5-((phenoxy- carbonyl)amino)-1- phenyl-1H-pyrazol-3- yl)pyrrolidine-1- carboxylate
MS (apci) m/z = 463.2 (M + H)





224


embedded image


(R)-tert-butyl 2-(5- ((phenoxycarbonyl) amino)-1-phenyl-1H- pyrazol-3- yl)pyrrolidine-1- carboxylate
MS (apci) m/z = 449.2 (M + H)





225


embedded image


(S)-tert-butyl 2-(5- ((phenoxycarbonyl) amino)-1-phenyl-1H- pyrazol-3- yl)pyrrolidine-1- carboxylate
MS (apci) m/z = 449.2 (M + H)





226


embedded image


tert-butyl 4-((5- ((phenoxycarbonyl) amino)-1-phenyl-1H- pyrazol-3- yl)methoxy)piperidine- 1-carboxylate
MS (apci) m/z = 493.2 (M + H)





227


embedded image


phenyl (3- (hydroxymethyl)-1- phenyl-1H-pyrazol-5- yl)carbamate
MS (apci) m/z = 310.1 (M + H)









Intermediate 228



embedded image


tert-butyl 4-((4-chloro-5-((phenoxycarbonyl)amino)-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate

To a suspension of tert-butyl 4-((5-(phenoxycarbonylamino)-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate (Intermediate 226), 98.5 mg, 0.200 mmol) in DCM (2.0 mL) was added pyridinium 4-methylbenzenesulfonate (PPTS) (5.03 mg, 0.020 mmol) and N-chlorosuccinimide (40.1 mg, 0.300 mmol). The resulting solution was stirred at ambient temperature for 8 days. The mixture was diluted with water and CH2Cl2, the organic layer was separated and the aqueous was extracted with CH2Cl2 (2×). The combined organic fractions were dried over MgSO4, filtered and concentrated. The residue was purified by silica chromatography using 30-40% EtOAc/hexanes gradient elution to afford the title compound as an orange oil (73.5 mg, 70% yield). MS (apci) m/z=527.2 (M+H).


Intermediate 229



embedded image


Phenyl (4-chloro-3-(hydroxymethyl)-1-phenyl-1H-pyrazol-5-yl)carbamate

Prepared from phenyl 3-(hydroxymethyl)-1-phenyl-1H-pyrazol-5-ylcarbamate (Intermediate 227) using the procedure outlined for the preparation of tert-butyl 4-((4-chloro-5-((phenoxycarbonyl)amino)-1-phenyl-1H-pyrazol-3-yl)methoxy)piperidine-1-carboxylate (Intermediate 228). In this instance, the compound was isolated a white solid (108 mg, 28%). MS (apci) m/z=344.0 (M+H).


Intermediate 230



embedded image


Phenyl (4-bromo-3-(hydroxymethyl)-1-phenyl-1H-pyrazol-5-yl)carbamate

To a suspension of phenyl 3-(hydroxymethyl)-1-phenyl-1H-pyrazol-5-ylcarbamate (Intermediate 227, 100 mg, 0.323 mmol) in CH2Cl2 (1.6 mL) was added pyridinium 4-methylbenzenesulfonate (PPTS) (8.12 mg, 0.0323 mmol) and N-bromosuccinimide (86.3 mg, 0.485 mmol). The reaction mixture was stirred for 16 hours at ambient temperature. The resulting suspension was filtered and the collected solid washed briefly with CH2Cl2 and dried in vacuum to afford the title compound a white solid (48.5 mg, 39%). MS (apci) m/z=388.0 (M+H).


The following pyrazole intermediates were made according to the methods described for the preparation of Intermediate 228, 229 or 230.


















MS (apci)


Intermediate
Structure
Name
m/z







231


embedded image


phenyl (4-chloro-3- (methoxymethyl)-1-phenyl- 1H-pyrazol-5-yl)carbamate
358.1 (M + H)





232


embedded image


phenyl (4-bromo-3- (methoxymethyl)-1-phenyl- 1H-pyrazol-5-yl)carbamate
402.2 (M + H)





233


embedded image


phenyl (4-chloro-3-(1,1- difluoro-2-hydroxyethyl)-1- phenyl-1H-pyrazol-5- yl)carbamate
394.1 (M + H)





234


embedded image


phenyl (4-chloro-3-(2- hydroxy-2-methylpropyl)-1- phenyl-1H-pyrazol-5- yl)carbamate
386.1 (M + H)





235


embedded image


(S)-phenyl (4-chloro-3-(2- hydroxypropyl)-1-phenyl-1H- pyrazol-5-yl)carbamate
372.1 (M + H)





236


embedded image


(R)-phenyl (4-chloro-3-(2- hydroxypropyl)-1-phenyl-1H- pyrazol-5-yl)carbamate
372.1 (M + H)





237


embedded image


(R)-phenyl (4-bromo-3-(2- hydroxypropyl)-1-phenyl-1H- pyrazol-5-yl)carbamate
416.0 (M + H)





238


embedded image


phenyl (4-chloro-3-(3- hydroxycyclobutyl)-1-phenyl- 1H-pyrazol-5-yl)carbamate
384.1 (M + H)





239


embedded image


phenyl 4-chloro-3-(3-methyl- 1,2,4-oxadiazol-5-yl)-1- phenyl-1H-pyrazol-5- ylcarbamate
396.0 (M + H)





240


embedded image


phenyl (4-chloro-3-(2- hydroxy-2-methylpropoxy)-1- phenyl-1H-pyrazol-5- yl)carbamate
446.1 (M + H)





241


embedded image


phenyl (4-chloro-3-(2- hydroxy-2-methylpropoxy)-1- phenyl-1H-pyrazol-5- yl)carbamate
388.1 (M + H)





242


embedded image


phenyl (4-bromo-3-(2- hydroxy-2-methylpropoxy)-1- phenyl-1H-pyrazol-5- yl)carbamate
433.0 (M + H)





243


embedded image


ethyl 4-bromo-5- ((phenoxycarbonyl)amino)-1- phenyl-1H-pyrazole-3- carboxylate
430.0 (M + H)









Intermediate 245



embedded image


5-methyl-3-phenyl-1-(pyrazin-2-yl)-1H-pyrazol-4-amine
Step A: 2-(5-methyl-4-nitroso-3-phenyl-1H-pyrazol-1-yl)pyrazine

To a solution of 2-hydrazinylpyrazine (0.485 g, 4.40 mmol) in HOAc (6 mL) was added (2-(hydroxyimino)-1-phenylbutane-1,3-dione (0.765 g, 4.00 mmol) in small portions over 2 minutes. The mixture was stirred for 5 minutes and the resulting light orange suspension was stirred at 60° C. for 6 hours. EtOH (1 mL) was added and the mixture was heated at 60° C. for an additional 6 hours. The resulting dark green suspension was cooled to ambient temperature and the mixture was diluted with H2O (30 mL). The green suspension was stirred for 1 hour and the solid was collected via vacuum filtration. The collected solid was washed with H2O and dried in vacuum. The solid was suspended in EtOH (25 mL) and concentrated HCl (500 μL) was added. The mixture was heated at reflux for 20 hours, cooled to ambient temperature and diluted with chilled H2O (75 mL). The mixture was treated with 1M NaOH to pH=7 and was extracted with Et2O (3×). The combined extracts were washed with saturated NaCl and dried over MgSO4. The dried solution was filtered through packed Celite® and concentrated. The residual green-yellow solid was purified on a SiO2 column using step gradient elution (25% CH2Cl2, 50% EtOAc/hexanes) to furnish the title compound as a turquoise solid (325 mg, 31%). MS (apci) m/z=266.1 (M+H).


Step B: 5-methyl-3-phenyl-1-(pyrazin-2-yl)-1H-pyrazol-4-amine

To a mixture of 2-(5-methyl-4-nitroso-3-phenyl-1H-pyrazol-1-yl)pyrazine (325 mg, 1.04 mmol) and Zn dust (340 mg, 5.21 mmol) in EtOH (10 mL) was added concentrated HCl (95.5 μL, 1.15 mmol). The mixture was stirred at ambient temperature for 17 hours, then at 65° C. for 3 hours. The mixture was cooled to ambient temperature and was filtered through packed Celite® eluting with MeOH. The eluent was concentrated, and the residue was treated with H2O and mixed. The resulting orange suspension treated with 2M HCl to pH=1 and the mixture was extracted with Et2O (3×). The aqueous portion was treated with 2M NaOH to pH=8 and extracted with EtOAc (3×). The combined EtOAc extracts were washed with saturated NaCl and dried over MgSO4/activated carbon. The solution was eluted through a SiO2 plug eluting with EtOAc. The eluent was concentrated to give the title compound as a light yellow wax (33 mg, 13%). MS (esi) m/z=252.2 (M+H).


Intermediate 246



embedded image


1,5-dimethyl-3-phenyl-1H-pyrazol-4-amine
Step A: 1,5-dimethyl-4-nitroso-3-phenyl-1H-pyrazole

To a solution of methylhydrazine (0.484 g, 10.5 mmol) in HOAc (10 mL) was added 2-(hydroxyimino)-1-phenylbutane-1,3-dione (2.01 g, 10.5 mmol) in small portions over 5 minutes. The reaction mixture was heated at 60° C. for 1 hour and was cooled to ambient temperature. Et2O (50 mL) and H2O (10 mL) were added to the mixture followed by slow addition of saturated Na2CO3 until pH=8 was obtained. The organic layer was removed and the aqueous layer was extracted with Et2O (2×). The combined organic fractions were dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography (1:5 EtOAc/hexanes) to give the title compound as a green solid (1.32 g, 63%). MS (apci) m/z=202.1 (M+H).


Step B: 1,5-dimethyl-3-phenyl-1H-pyrazol-4-amine

To a solution of 1,5-dimethyl-4-nitroso-3-phenyl-1H-pyrazole (1.32 g, 6.60 mmol) in MeOH (50 mL) was added Pd(OH)2 on carbon (200 mg, 20 wt %, 0.286 mmol) and the reaction mixture was shaken under 50 psi of H2 for 3 hours at ambient temperature. The reaction mixture was evacuated, purged with N2 filtered through a pad of Celite® with MeOH elution. The eluent was concentrated and the residue dried in vacuum to provide the title compound as a tan solid (1.23 g, 100%). MS (apci) m/z=188.1 (M+H).


Intermediate 247



embedded image


1-isopropyl-5-methyl-3-phenyl-1H-pyrazol-4-amine

The title compound was prepared according to the method described for Intermediate 246, using isopropylhydrazine hydrochloride in place of methylhydrazine in Step A to provide 620 mg (57%) of the title compound over 2 steps. MS (apci) m/z=216.1 (M+H).


Intermediate 248



embedded image


5-methyl-3-phenyl-1-(2,2,2-trifluoroethyl)-1H-pyrazol-4-amine
Step A: 5-methyl-4-nitroso-3-phenyl-1-(2,2,2-trifluoroethyl)-1H-pyrazole

The title compound was prepared using (2,2,2-trifluoroethyl)hydrazine in place of methylhydrazine in Step A of the procedure described for the preparation of 1,5-dimethyl-3-phenyl-1H-pyrazol-4-amine (Intermediate 246). The compound was isolated as a green solid (999 mg, 71%). 1H NMR (CDCl3) δ 7.60-7.73 (m, 5H), 4.70 (q, 2H), 2.27 (t, 3H).


Step B: 5-methyl-3-phenyl-1-(2,2,2-trifluoroethyl)-1H-pyrazol-4-amine

To a mixture of 5-methyl-4-nitroso-3-phenyl-1-(2,2,2-trifluoroethyl)-1H-pyrazole (50 mg, 0.186 mmol) and Zn dust (60.7 mg, 0.929 mmol) in EtOH (0.4 mL) was added concentrated HCl (17.0 μL, 0.204 mmol) and the mixture was heated at reflux for 3 hours. The mixture was cooled to ambient temperature and was diluted with MeOH and filtered. The filtrate was concentrated and the residue was diluted in water. The aqueous mixture was treated with saturated NaHCO3 until pH=10 was achieved. The mixture was extracted with DCM (3×) and the combined extracts were dried over Na2SO4, filtered and concentrated afford the title compound as a yellow oil (47.1 mg, 99.4% yield). MS (apci) m/z=256.1 (M+H).


Intermediate 249



embedded image


1-ethyl-5-methyl-3-phenyl-1H-pyrazol-4-amine
Step A: 1-ethyl-5-methyl-4-nitroso-3-phenyl-1H-pyrazole

The title compound was prepared according to the procedure described for the preparation of Intermediate 246, using ethylhydrazine oxalate in place of methylhydrazine in Step A. 1-Ethyl-5-methyl-4-nitroso-3-phenyl-1H-pyrazole was isolated as a green oil (288 mg, 26%). 1H NMR (CDCl3) δ 8.19 (d, 2H), 7.46-7.50 (m, 3H), 4.15 (q, 2H), 2.43 (s, 3H), 1.50 (t, 3H). The minor regioisomer, 1-ethyl-3-methyl-4-nitroso-5-phenyl-1H-pyrazole, was also obtained as a blue-green solid (165 mg, 15%). 1H NMR (CDCl3) δ 7.71 (dd, 2H), 7.59 (m, 3H), 4.17 (q, 2H), 2.28 (s, 3H), 1.51 (t, 3H).


Step B: 1-ethyl-5-methyl-3-phenyl-1H-pyrazol-4-amine

Prepared according to the procedure described for the preparation of Intermediate 248, using 1-ethyl-5-methyl-4-nitroso-3-phenyl-1H-pyrazole in Step B. the title compound was isolated as a light purple solid (281 mg, 104%). MS (apci) m/z=202.1 (M+H).


Intermediate 250



embedded image


1-ethyl-3-methyl-5-phenyl-1H-pyrazol-4-amine

Prepared according to the procedure described for the preparation of Intermediate 249, using 1-ethyl-3-methyl-4-nitroso-5-phenyl-1H-pyrazole in Step A. The title compound was prepared according to Step B. The compound was isolated as a colorless oil (82.4 mg, 52.5%) after purification by reverse-phase chromatography. MS (apci) m/z=202.1 (M+H).


Intermediate 251



embedded image


1-methyl-5-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-amine
Step A: 4,4,4-trifluoro-2-(hydroxyimino)-1-phenylbutane-1,3-dione

A solution of 4,4,4-trifluoro-1-phenylbutane-1,3-dione (5.00 g, 23.1 mmol) in HOAc (46.3 mL) was chilled to 10° C. and sodium nitrite (1.84 g, 26.6 mmol) in water (6.0 mL) was added. The mixture was stirred at ambient temperature for 90 minutes and was diluted with H2O (150 mL). The mixture was extracted with Et2O (3×) and the combined organic fractions were carefully washed with saturated NaHCO3 until pH=9. The Et2O solution was washed with H2O and saturated NaCl and was dried over MgSO4. The dried solution was filtered and concentrated to afford the title compound as a yellow foam (4.21 g, 74.2% yield). MS (apci) m/z=244.1 (M−H).


Step B: 4-nitroso-3-phenyl-5-(trifluoromethyl)-1H-pyrazole

A solution of hydrazine monohydrate (0.204 g, 4.08 mmol) in EtOH (5 mL) was cooled to 0° C. and 4,4,4-trifluoro-2-(hydroxyimino)-1-phenylbutane-1,3-dione (1.00 g, 4.08 mmol) in EtOH (15 mL) was added. The reaction mixture was stirred at ambient temperature for 3 hours, excess powdered MgSO4 was added and the mixture was heated at 60° C. for 16 hours. The mixture was cooled to ambient temperature, filtered and concentrated to afford the crude title compound as a green solid (78.7 mg, 8.0%) that was taken directly to the next step. MS (apci) m/z=240.0 (M−H).


Step C: 1-methyl-5-phenyl-3-(trifluoromethyl)-1H-pyrazol-4-amine

To a solution of 4-nitroso-3-phenyl-5-(trifluoromethyl)-1H-pyrazole (78.7 mg, 0.326 mmol) in DMF (1.6 mL) was added NaH (14.4 mg, 0.359 mmol) and the mixture was stirred at ambient temperature for 30 minutes. The mixture was treated with methyl iodide (40.6 μL, 0.653 mmol) and stirred for 17 hours. The reaction mixture was directly purified by reverse phase HPLC using 20-100% acetonitrile/water gradient elution to provide a light blue solid (40.2 mg). The solid was dissolved in EtOH (0.35 mL) and was subjected to the reduction procedure described in Step B of the preparation of 5-methyl-3-phenyl-1-(2,2,2-trifluoroethyl)-1H-pyrazol-4-amine (Intermediate 248). The title compound was obtained as white solid (25.1 mg, 66.1%).


Intermediate 252



embedded image


1-methyl-3-phenyl-5-(trifluoromethyl)-1H-pyrazol-4-amine
Step A: 1-methyl-4-nitroso-3-phenyl-5-(trifluoromethyl)-1H-pyrazole

To a solution of methylhydrazine (0.214 mL, 4.08 mmol) in EtOH (20 mL) was added 4,4,4-trifluoro-2-(hydroxyimino)-1-phenylbutane-1,3-dione (Intermediate 251, Step A; 1.00 g, 4.079 mmol). The reaction mixture was stirred at ambient temperature for 1 hour and excess MgSO4 was added. The mixture was stirred at 60° C. for 48 hours and was cooled to ambient temperature. The mixture was filtered and the filtrate concentrated to a green residue. The residue was purified by silica gel chromatography using a 10-30% EtOAc/hexanes gradient for elution to provide the title compound as a green solid (482 mg, 46%). 1H NMR (CDCl3) δ 7.89 (d, 2H), 7.45-7.52 (m, 3H), 4.15 (s, 3H).


Step B: 1-methyl-3-phenyl-5-(trifluoromethyl)-1H-pyrazol-4-amine

Prepared from 1-methyl-4-nitroso-3-phenyl-5-(trifluoromethyl)-1H-pyrazole according to the method described for the preparation of Intermediate 248, Step B. The title compound was obtained as white solid (309 mg, 68%). 1H NMR (CDCl3) δ 7.65 (d, 2H), 7.45 (t, 2H), 7.35 (t, 1H), 3.93 (s, 3H), 3.52 (br s, 2H).


Intermediate A



embedded image


phenyl (3-carbamoyl-4-methyl-1-phenyl-1H-pyrazol-5-yl)carbamate
Step A: 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbonitrile

To a stirred solution of aniline (2.02 g, 21.7 mmol) in 6 N HCl (22 mL) was added dropwise a solution of NaNO2 (1.50 g, 21.7 mmol) in water (20 mL) at 0-5° C. The reaction mixture was stirred at 0° C. for 15 minutes. Acetic acid (10 mL) was added. This solution was added dropwise to a stirred solution of ethyl 2,3-dicyanobutanoate (3.60 g, 21.7 mmol; Prepared according to the procedure described in Bioorganic & Medicinal Chemistry, 2004, 12, 3345-3356) in acetic acid (12 mL) and water (18 mL) at 0° C. After stirring for 1 hour, concentrated ammonium hydroxide (50 mL) was added dropwise followed by THF (50 mL). The reaction was stirred at ambient temperature overnight. The organic layer was separated. The aqueous layer was extracted with EtOAc. The combined organic layers were washed with brine, dried and concentrated. The residue was purified by flash chromatography on silica gel (3:1 hexanes/EtOAc) to give the title compound (2.95 g, 69% yield). MS (apci) m/z=198.9 (M+H).


Step B: 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxamide

A solution of 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carbonitrile (150 mg, 0.757 mmol) in concentrated H2SO4 (0.5 mL) was stirred at ambient temperature for 17 hours. The reaction mixture was cooled and neutralized by the addition of aqueous NaOH (2M, 11 mL), then extracted 10% MeOH/DCM (5×10 mL), and the combined organic extracts were washed with brine, dried (MgSO4), filtered and concentrated under reduced pressure to afford the title compound as a white solid (151 mg, 95% yield). MS (apci) m/z=239.1 (M+Na).


Step C: phenyl (3-carbamoyl-4-methyl-1-phenyl-1H-pyrazol-5-yl)carbamate

To a solution of 5-amino-4-methyl-1-phenyl-1H-pyrazole-3-carboxamide (163 mg, 0.754 mmol) in EtOAc (7 mL) was added sodium hydroxide (2M aq, 0.754 mL, 1.508 mmol) followed by addition of phenyl carbonochloridate (0.104 mL, 0.829 mmol). The reaction mixture was stirred at ambient temperature for 21 hours and then was transferred to a separatory funnel. The phases were separated. The organic phase was washed with H2O and brine, dried over MgSO4, filtered and concentrated to afford the product as a pale yellow thick syrup (284 mg, 112% yield), which was used without further purification. MS (apci) m/z=337.1 (M+H).


Intermediate B



embedded image


3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine
Step A: 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one

A mixture of ethyl 2-cyanopropanoate (50.5 g, 397.2 mmol) and phenylhydrazine (39 mL, 397.2 mmol) in dioxane (100 mL) was heated at 110° C. for 5 days. The cooled mixture was concentrated to ½ volume and then cooled in ice and triturated with cold Et2O. The resulting solids were filtered, washed extensively with Et2O and dried under vacuum to afford 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (34.69 g, 46% yield) as a fluffy white powder. MS (apci) m/z=190.1 (M+H).


Step B: 3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine

To a solution of 5-amino-4-methyl-1-phenyl-1H-pyrazol-3(2H)-one (500 mg, 2.643 mmol) in DMF (5 mL) were added K2CO3 (730 mg, 5.285 mmol) and (2-bromoethoxy)(tert-butyl)dimethylsilane (1.134 mL, 5.285 mmol). The reaction mixture was heated to 60° C. for 17 hours, then cooled to ambient temperature. The reaction mixture was filtered, diluted with EtOAc (60 mL), washed with water and brine, dried (MgSO4), filtered and concentrated. The crude product was purified by silica column chromatography eluting with 0-40% acetone/hexane, to afford the title compound (388 mg, 42% yield) as a waxy off-white solid. MS (apci) m/z=348.2 (M+H).


SYNTHETIC EXAMPLES
Example 1



embedded image


1-(1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)-3-(2-methyl-4-phenylthiazol-5-yl)urea
Step A: Preparation of N-(cyano(phenyl)methyl)acetamide

To a solution of 2-amino-2-phenylacetonitrile (1.00 g, 7.566 mmol) in THF (75 mL) were added DIEA (1.98 mL, 11.35 mmol) then acetyl chloride (0.592 mL, 8.323 mmol). The reaction mixture was stirred at ambient temperature for 20 hours, then concentrated, diluted with water (50 mL) and extracted with DCM (3×50 mL). The combined organic phases were washed with brine (2×100 mL), dried over MgSO4, filtered and concentrated to afford the product as a tan solid (1.216 g, 92% yield). 1H NMR (CDCl3) δ 7.48 (m, 2H), 7.43 (m, 3H), 6.14 (d, 1H), 6.02 (br s, 1H), 2.08 (s, 3H).


Step B: Preparation of 2-methyl-4-phenylthiazol-5-amine

To N-(cyano(phenyl)methyl)acetamide (150 mg, 0.861 mmol) were added Lawesson's Reagent (383 mg, 0.947 mmol) and toluene (2 mL). The reaction mixture was heated to reflux for 44 hours, then purified by reverse-phase column chromatography, eluting with 5-85% acetonitrile/water, to afford the title compound as a yellow solid (14.9 mg, 9% yield). MS (apci) m/z=191.1 (M+H).


Step C: Preparation of 1-(1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)-3-(2-methyl-4-phenylthiazol-5-yl)urea

To a solution of 2-methyl-4-phenylthiazol-5-amine (14.9 mg, 0.078 mmol) in DMF (0.6 mL) were added DIEA (27 μL, 0.157 mmol) then phenyl (1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)carbamate (Intermediate 2, 29.2 mg, 0.0783 mmol). The reaction mixture was heated to 60° C. for 18 hours, then was diluted with water (25 mL) and extracted with EtOAc (2×25 mL). The combined organic layers were rinsed with brine (2×50 mL), dried over MgSO4, filtered, and concentrated. The crude product was purified by reverse-phase column chromatography, eluting with 5-85% acetonitrile/water, to afford the title compound as a white solid (13.2 mg, 36% yield). MS (apci) m/z=470.1 (M+H).


Example 2



embedded image


1-(1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea
Step A: Preparation of N-(cyano(phenyl)methyl)-2,2,2-trifluoroacetamide

To a mixture of 2-amino-2-phenylacetonitrile hydrochloride (4.00 g, 20.16 mmol) and DIEA (10.54 mL, 60.49 mmol) in DCM (120 mL) cooled to 0° C. was added TFAA (3.083 mL, 22.18 mmol) dropwise over 5 minutes. The reaction mixture was stirred at ambient temperature for 30 minutes, then diluted with water (100 mL), phases separated and the aqueous phase extracted with DCM (2×50 mL). The combined organic phases were washed with brine (100 mL), dried over MgSO4, filtered and concentrated. The crude product was purified by silica column chromatography, eluting with 0-40% acetone in hexanes, to afford the product as a yellow solid (1.56 g, 34% yield). 1H NMR (CDCl3) δ 7.50 (m, 5H), 6.83 (br s, 1H), 6.08 (d, 1H).


Step B: Preparation of 1-(1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea

Prepared according to the procedure of Example 1, Steps B-C, replacing N-(cyano(phenyl)methyl)acetamide with N-(cyano(phenyl)methyl)-2,2,2-trifluoroacetamide in Step B. The crude product was purified by reverse-phase column chromatography, eluting with 5-85% acetonitrile/water, to afford the title compound as a white solid (7.2 mg, 9% yield). MS (apci) m/z=524.2 (M+H).


Example 3



embedded image


1-(1′,4-dimethyl-1-phenyl-1H, 1′H-[3,4′-bipyrazol]-5-yl)-3-(2-methyl-4-phenyloxazol-5-yl)urea
Step A: Preparation of 2-methyl-4-phenyloxazol-5-amine

To a solution of N-(cyano(phenyl)methyl)acetamide (Example 1, Step A, 200 mg, 1.15 mmol) in dioxane (1.5 mL) was added HCl (4.0 M in dioxane, 1.15 ml, 4.59 mmol). The reaction mixture was stirred at ambient temperature for 24 hours, then diluted with 1M aq. NaOH (25 mL) and extracted with EtOAc (3×25 mL). The combined organic layers were washed with brine (50 mL), dried over MgSO4, filtered, and concentrated to afford the product as a brown oil (168 mg, 84% yield). MS (apci) m/z=175.1 (M+H).


Step B: Preparation of 1-(1′,4-dimethyl-1-phenyl-1H,1′H-[3,4′-bipyrazol]-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea

Prepared according to the procedure of Example 1, Step C, replacing 2-methyl-4-phenylthiazol-5-amine with 2-methyl-4-phenyloxazol-5-amine. The crude product was purified by reverse-phase column chromatography, eluting with 5-85% acetonitrile/water, to afford the title compound as a white solid (4.0 mg, 6% yield). MS (apci) m/z=454.2 (M+H).


Example 4



embedded image


4-methyl-1-phenyl-5-(3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)ureido)-1H-pyrazole-3-carboxamide
Step A: 4-phenyl-2-(trifluoromethyl)thiazol-5-amine

In a 150-mL pressure tube to solid N-(cyano(phenyl)methyl)-2,2,2-trifluoroacetamide (Example 2, Step A, 1.46 g, 6.40 mmol) were added Lawesson's Reagent (2.64 g, 6.53 mmol) and toluene (20 mL). The reaction mixture was heated to 100° C. for 24 hours, then cooled to ambient temperature. The reaction mixture was purified by silica column chromatography eluting with 0-40% acetone/hexane, to afford the title compound (479 mg, 31% yield) as a yellow solid. MS (apci) m/z=245.1 (M+H).


Step B: 4-methyl-1-phenyl-5-(3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)ureido)-1H-pyrazole-3-carboxamide

To a mixture of phenyl (3-carbamoyl-4-methyl-1-phenyl-1H-pyrazol-5-yl)carbamate (Intermediate A, 50 mg, 0.15 mmol) and 4-phenyl-2-(trifluoromethyl)thiazol-5-amine (36 mg, 0.15 mmol) in DMF (0.6 mL) was added DIEA (0.052 mL, 0.30 mmol). The reaction mixture was stirred at ambient temperature for 18 hours. The mixture was purified by reverse-phase column chromatography, eluting with 5-90% acetonitrile/water with 0.1% TFA, to afford the title compound as a pale yellow solid (2.5 mg, 4% yield). MS (apci) m/z=487.1 (M+H).


Example 5



embedded image


1-(3-(2-hydroxyethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea
Step A: phenyl (4-phenyl-2-(trifluoromethyl)thiazol-5-yl)carbamate

To a solution of 4-phenyl-2-(trifluoromethyl)thiazol-5-amine (Example 4, Step A, 100 mg, 0.409 mmol) in EtOAc (4 mL) were added aqueous NaOH (2M, 0.409 mL, 0.819 mmol) and phenylchloroformate (0.062 mL, 0.491 mmol). The reaction mixture was stirred at ambient temperature for 23 hours. The reaction mixture was transferred to a separatory funnel with 10 mL EtOAc. The phases were separated, and the organic phase was washed with H2O and brine, dried (MgSO4), filtered and concentrated to a yellow solid. The crude solid was purified by silica column chromatography eluting with 0-40% acetone/hexane, to afford the title compound (121 mg, 81% yield) as a yellow solid. MS (apci) m/z=365.0 (M+H).


Step B: 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea

To phenyl (4-phenyl-2-(trifluoromethyl)thiazol-5-yl)carbamate (30 mg, 0.082 mmol) and 3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-amine (Intermediate B, 29 mg, 0.082 mmol) was added i-PrOH (0.8 mL). The reaction mixture was heated to 50° C. for 24 hours. The reaction mixture was purified by silica column chromatography eluting with 0-30% acetone/hexane, to afford the title compound (6.8 mg, 13% yield) as a yellow solid. MS (apci) m/z=618.2 (M+H).


Step C: 1-(3-(2-hydroxyethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea

To a solution of 1-(3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-4-methyl-1-phenyl-1H-pyrazol-5-yl)-3-(4-phenyl-2-(trifluoromethyl)thiazol-5-yl)urea (6.8 mg, 0.011 mmol) in EtOH (1.5 mL) was added HCl (5-6 M in i-PrOH, 0.005 mL). The reaction mixture was stirred at ambient temperature for 1.5 hours, then concentrated. The solid was diluted with Et2O (2×1 mL) and concentrated after each addition. The crude product was purified by silica column chromatography eluting with 1% NH3/9% MeOH/90% DCM, to give the title compound as a pale yellow solid (5.7 mg, 103% yield). MS (apci) m/z=504.1 (M+H).

Claims
  • 1. A compound of Formula I:
  • 2. The compound according to claim 1, wherein R4 is pyrazolyl optionally substituted with one or more groups independently selected from (1-6C)alkyl.
  • 3. A compound according to claim 1, selected from the group consisting of
  • 4. A pharmaceutical composition, which comprises a compound of Formula I as defined in claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable diluent or carrier.
RELATED APPLICATIONS

This application is a 371 filing of PCT Application No. PCT/US2013/069728 filed Nov. 12, 2013, which claims priority to U.S. Provisional Application Ser. No. 61/725,950, filed Nov. 13, 2012, each of which is incorporated herein by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2013/069728 11/12/2013 WO 00
Publishing Document Publishing Date Country Kind
WO2014/078322 5/22/2014 WO A
US Referenced Citations (8)
Number Name Date Kind
5849779 Hirota et al. Dec 1998 A
5998424 Galemmo, Jr. et al. Dec 1999 A
6197798 Fink et al. Mar 2001 B1
6410533 Hirth et al. Jun 2002 B1
7223782 Atkinson et al. May 2007 B2
7625915 Dumas et al. Dec 2009 B2
8592454 Shirai et al. Nov 2013 B2
9163017 Degoey Oct 2015 B2
Foreign Referenced Citations (75)
Number Date Country
0761658 Dec 1997 EP
1043995 Nov 2006 EP
2033955 Mar 2009 EP
1451160 Jan 2010 EP
2336105 Sep 2014 EP
2005206527 Aug 2005 JP
9804521 Feb 1998 WO
9923091 May 1999 WO
9932110 Jul 1999 WO
0039116 Jul 2000 WO
0043384 Jul 2000 WO
200112188 Feb 2001 WO
200202525 Jan 2002 WO
2002088101 Nov 2002 WO
2002090326 Nov 2002 WO
2003037274 May 2003 WO
2003045920 Jun 2003 WO
2003051275 Jun 2003 WO
2004005262 Jan 2004 WO
2004032870 Apr 2004 WO
2004060305 Jul 2004 WO
2004060306 Jul 2004 WO
2004061084 Jul 2004 WO
2004111009 Dec 2004 WO
2005024755 Mar 2005 WO
2005048948 Jun 2005 WO
2005110994 Nov 2005 WO
2006014290 Feb 2006 WO
2006068591 Jun 2006 WO
2006070198 Jul 2006 WO
2006071940 Jul 2006 WO
2006081034 Aug 2006 WO
2006123257 Nov 2006 WO
2007008917 Jan 2007 WO
2007059202 May 2007 WO
2007061882 May 2007 WO
2007064872 Jun 2007 WO
2008016811 Feb 2008 WO
2008021859 Feb 2008 WO
2008033999 Mar 2008 WO
2008034008 Mar 2008 WO
2008046003 Apr 2008 WO
2008131276 Oct 2008 WO
2008150899 Dec 2008 WO
2009140128 Nov 2009 WO
2010032856 Mar 2010 WO
2010033941 Mar 2010 WO
2010040663 Apr 2010 WO
2010048314 Apr 2010 WO
2010059719 May 2010 WO
2010075376 Jul 2010 WO
2010077680 Jul 2010 WO
2010104488 Sep 2010 WO
2010125799 Nov 2010 WO
2011006074 Jan 2011 WO
2011032291 Mar 2011 WO
2011146336 Nov 2011 WO
2012158413 Nov 2012 WO
2013063214 May 2013 WO
2013096226 Jun 2013 WO
2013176970 Nov 2013 WO
2014052563 Apr 2014 WO
2014052566 Apr 2014 WO
2014078322 May 2014 WO
2014078323 May 2014 WO
2014078325 May 2014 WO
2014078328 May 2014 WO
2014078331 May 2014 WO
2014078372 May 2014 WO
2014078378 May 2014 WO
2014078408 May 2014 WO
2014078417 May 2014 WO
2014078454 May 2014 WO
2015039333 Mar 2015 WO
2015042085 Mar 2015 WO
Non-Patent Literature Citations (56)
Entry
Tsuzuki, Y., et al., Tetrahedron Asymmetry 12 (2001), 2989-2997.
Wadhwa, S., et al., Journal of Biosciences, 2003, 28(2), 181-188.
Wang, T., et al., Expert Opinion in Therapeutic Patents (2009) 19(3):305-319.
Woolf, C.J. et al. (1994) Neuroscience, 62, 327-331.
Yilmaz, T., et al., Cancer Biology and Therapy, 2010, 10(6), 644-653.
Zahn, P.K. et al. (2004) J. Pain, 5, 157-163.
Patent Cooperation Treaty, International Searching Authority, Search Report and Written Opinion for PCT/US2013/069728, dated May 28, 2015, 8 Pages.
Adriaenssens, E., et al. Cancer Res (2008) 68:(2) 346-351.
Asaumi, K., et al., Bone (2000) 26(6) 625-633.
Bardelli, A., Science 2003, 300, 949.
Bhattacharya, S. K., et al., Bioorganic & Medicinal Chemistry Letters (2012) 22(24) 7523-7592.
Bouhana, Karyn S., et al., “Comparison of Analgesic Effects of an Allosteric Inhibitor of TrkA to that of an ATP site inhibitor of the pan-Trk axis in a Rodent Model of Inflammatory Pain”, Gordon Conference, Salve Regina University, Newport, RI, Jun. 7, 2011.
Brodeur, G. M., Nat. Rev. Cancer 2003, 3, 203-216.
Bruno, O., Bioorganic & Medicinal Chemistry (2009) 17, 3379-3387.
Burger, K., et al., Synthesis (1990) vol. 4, 360-365.
Chambers, L. J., et al., Bioorganic & Medicinal Chemistry Letters (2010) 20(10) 3161-3164.
Davidson. B., et al., Clin. Cancer Res. 2003, 9, 2248-2259.
Davies, Stephen G., et al., Asymmetric synthesis of 3,4-anti- and 3,4-syn-substituted aminopyrrolidines via lithium amide conjugate addition, Org. Biomol. Chem., 2007, 5, 1961-1969.
Delafoy, L. et al. (2003) Pain 105, 489-497.
Demelo-Jorge, M. et al., Cell Host & Microbe (2007) 1(4), 251-261.
Dimola, F. F, et. al., Gut (2000) 46(5), 670-678.
Dou, Y.-C., et. al. Archives of Dermatological Research (2006) 298(1), 31-37.
Du, et al., World Journal of Gastroenterology, 2003, 9(7), 1431-1434.
Eguchi, M., et al., Blood 1999, 93 (4), pp. 1355-1363.
El Haddad, M., et al., J. Heterocyclic Chem., (2000) 37, 1247-1252.
Eliav, E. et al., Pain 79, 255-264 (1999).
Euthus, D.M., et al., Cancer Cell 2002, 2 (5), pp. 347-348.
Freund-Michel, V; Frossard, N., Pharmacology & Therapeutics (2008) 117(1), 52-76.
Greco, A., et al., Molecular and Cellular Endocrinology 2010, 321 (1), pp. 44-49.
Gruber-Olipitz, M., et al., Journal of Proteome Research 2008, 7 (5), pp. 1932-1944.
Gwak, Y. S. et al. (2003) Neurosci. Lett. 336, 117-120.
Han, S., et al., J. Biological Chem., (2009), 284(19) 13199-13201.
Herzberg, U. et al., Neuroreport 1997; 8:1613-1618.
Hu, Vivian Y; et al., The Journal of Urology (2005), 173(3), 1016-1021.
Jaggar, S. I. et al., Br. J. Anaesth. (1999) 83, 442-448.
Jin, W., et al., Carcinogenesis (2010) 31 (11), pp. 1939-1947.
Kaymakcioglu, B.K., et al., European Journal of Pharmaceutical Sciences (2005) 26(1), 97-103.
Lamb, K. et al. (2003) Neurogastroenterol. Motil. 15, 355-361.
Li, L. et al. (2003) Mol. Cell. Neurosci. 23, 232-250.
Li, Y.-G., et al., Chinese Journal of Cancer Prevention and Treatment, 2009, 16 (6), pp. 428-430 (with English Abstract).
Ma, Q. P. and Woolf, C. J. NeuroReport (1997) 8, 807-810.
Mantyh, Patrick W., et al., Anesthesiology, vol. 115, No. 1, Jul. 2011, 189-204.
McCarthy, C. and Walker, E., Expert Opin. Ther. Patents (2014) 24(7):731-744.
McMahon, S.B. et al., (1995) Nat. Med. 1, 774-780.
Meyer, J. et al. (2007) Leukemia, 21(10):2171-2180.
Nakagawara, A. (2001) Cancer Letters 169:107-114.
Patapoutian, A. et al., Current Opinion in Neurobiology, 2001, 11, 272-280.
Pierottia, M.A. and Greco A., (2006) Cancer Letters 232:90-98.
Pinski, J. et al., Cancer Research, (2002) 62:986-989.
Ramer, M. S. and Bisby, M. A. (1999) Eur. J. Neurosci. 11, 837-846.
Raychaudhuri, S. P., et al., J. Investigative Dermatology (2004) 122(3), 812-819.
Ricci A., et al., American Journal of Respiratory Cell and Molecular Biology, 2001, 25(4), pp. 439-446.
Ro, L. S. et al., Pain, Feb. 1999; 79(2-3):265-274.
Shelton, D. L. et al. (2005) Pain, 116, 8-16.
Theodosiou, M. et al. (1999) Pain, 81, 245-255.
Truzzi, F., et al., Dermato-Endocrinology, 2011, 3(1), 32-36.
Related Publications (1)
Number Date Country
20160280702 A1 Sep 2016 US
Provisional Applications (1)
Number Date Country
61725950 Nov 2012 US