WO2015123133A1 - Pyrazolone compounds and uses thereof - Google Patents

Pyrazolone compounds and uses thereof Download PDF

Info

Publication number
WO2015123133A1
WO2015123133A1 PCT/US2015/015000 US2015015000W WO2015123133A1 WO 2015123133 A1 WO2015123133 A1 WO 2015123133A1 US 2015015000 W US2015015000 W US 2015015000W WO 2015123133 A1 WO2015123133 A1 WO 2015123133A1
Authority
WO
WIPO (PCT)
Prior art keywords
propyl
oxo
phenoxy
phenyl
dimethyl
Prior art date
Application number
PCT/US2015/015000
Other languages
French (fr)
Inventor
Nicholas Simon Stock
Austin Chih-Yu Chen
Yalda Mostofi BRAVO
Jason Duarte JACINTHO
Yen TROUNG
Original Assignee
Inception 2, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=53800550&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2015123133(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to KR1020167025019A priority Critical patent/KR20160115996A/en
Priority to AU2015217450A priority patent/AU2015217450A1/en
Priority to CA2937542A priority patent/CA2937542A1/en
Priority to US15/118,402 priority patent/US9862708B2/en
Priority to CN201580008005.1A priority patent/CN106029640B/en
Priority to MX2016010436A priority patent/MX2016010436A/en
Priority to NZ722345A priority patent/NZ722345A/en
Priority to EP15748977.4A priority patent/EP3105205A4/en
Priority to SG11201606149YA priority patent/SG11201606149YA/en
Priority to EA201691342A priority patent/EA201691342A1/en
Application filed by Inception 2, Inc. filed Critical Inception 2, Inc.
Priority to JP2016552317A priority patent/JP2017505804A/en
Priority to BR112016017781A priority patent/BR112016017781A2/en
Publication of WO2015123133A1 publication Critical patent/WO2015123133A1/en
Priority to ZA2016/05222A priority patent/ZA201605222B/en
Priority to IL247148A priority patent/IL247148A0/en
Priority to US15/829,514 priority patent/US20180079745A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41521,2-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. antipyrine, phenylbutazone, sulfinpyrazone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • AIDS & HIV (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The invention disclosed herein is directed to compounds of Formula I [Formula should be entered here] and pharmaceutically acceptable salts thereof, which are useful in the treatment of prostate, breast, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma, and other cancers. The invention also includes pharmaceutical compositions comprising a therapeutically effective amount of compound of Formula I, or a pharmaceutically acceptable salt thereof. The invention disclosed herein is also directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers. The invention disclosed herein is further directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers through the administration of a therapeutically effective amount of a selective PPARα antagonist. The compounds and pharmaceutical compositions of the invention are also useful in the treatment of viral infections, such as HCV infections and HIV infections.

Description

PYRAZOLONE COMPOUNDS AND USES THEREOF
FIELD OF THE INVENTION
[0001] This invention is directed to novel pyrazolones, or pharmaceutically acceptable salts thereof, useful in the treatment of prostate, breast, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers. The invention disclosed herein is further directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers comprising administration of selective PPARa antagonists. The compounds and pharmaceutical compositions of the invention are also useful in the treatment of viral infections, such as HCV infections and HIV infections.
BACKGROUND OF THE INVENTION
[0002] While tremendous strides have been made in the treatment of various cancers, in many cases, cancer treatment continues to be a matter of administering one or more anti-cancer agents that are marginally less chemotoxic to healthy cells than they are to the cancer in question. In recognition of this problem, there has been substantial research effort aimed at identifying, understanding and taking advantage of phenotypical behavior peculiar to certain cancer cells. It has long been observed that most cancer cell types generate energy for cellular processes through aerobic glycolysis rather than through oxidative phosphorylation as found in the normal cell. This process, which became known as the "Warburg effect", is highly energy inefficient and requires cancer cell mitochondria to resort to glucose fermentation to make up the energy deficit. Since perhaps the mid- 1990 's researchers have sought to identify methods of treating cancer that take advantage of the "Warburg effect" and associated aspects of cancer cell mitochondrial metabolism. See, for example, Wang, et. al , Small mitochondrial-targeting molecules as anti-cancer agents, Mol. Aspects Med. 2010 February; 31 (1 ): 75-92.
[0003] Samudio, et. al , J. Clin. Invest. 120: 142-156 (2010), disclosed that in certain leukemia cell lines "mitochondrial uncoupling - the continuing reduction of oxygen without ATP synthesis - has recently been shown in leukemic cells to circumvent the ability of oxygen to inhibit glycolysis, and may promote the metabolic preference for glycolysis by shifting from pyruvate oxidation to fatty acid oxidation (FAO)." Samudio, et. al , also provided data indicating that inhibition of FAO could sensitize human leukemia cells to apoptosis, and further that inhibition of FAO may prove useful in the treatment of leukemia.
[0004] PPARa is known to be an important regulator of fatty acid oxidation. See Pyper, et. al, Nucl. Recept. Signal. 8:e002., e002 (2010). It has been reported that expression of the PPARa gene can be higher in human chronic lymphocytic leukemia (CLL) making this cancer type sensitive to therapies aimed at reducing FAO (Samudio et al, J. Clin. Invest. 120: 142-156 (2010)). This effect may generalize to several cancer types. For example, ovarian cancer and breast cancer (Linher-Melville et al , 201 1 , BMC, 4; 1 1 :56), thrive in an adipose rich environment and as a result can be negatively impacted by targeted therapies that reduce fatty acid metabolism (Nieman et al , 201 1 , Nat Med. 201 1 Oct 30; 17(1 1 ): 498-503). Still other cancers that rely on FAO include prostate cancer (Liu, Prostate Cancer Prostatic Dis., 2006; 9(3):230-4), colon cancer (Holla et ah, 201 1 , JCB 286(34):30003-30009), pancreatic cancer (Khasawneh et ah , 2009, PNAS, 106(9):3354-3359) and lung cancer (Zaugg et ah , 201 1 , Genes and Development, 25: 1041 -1051 ).
[0005] GW6471 (Xu, et. ah , Nature 415, 813-817 (2002)) and MK-866 (Kehrer, et. ah , Biochem. J. 356, 899-906 (2001)) have been identified as antagonists of PPARa. Moreover, MK-866, whose primary activity is as an inhibitor of FLAP, has been disclosed to induce apoptosis in a human chronic lymphocytic leukemia cell line in a FLAP-independent manner; and has also been disclosed to induce apoptosis in prostate and glioblastoma cell lines.
[0006] Chronic myeloid leukemia (CML) is a model of hematopoietic stem cell (HSC) disease. In 2008, Ito et al, disclosed evidence linking the loss of promyelocytic leukemia (PML) gene expression with favorable outcomes in CML (Nature, 2008 June 19; 453 (7198) 1072-1078). More recently Ito et ah, disclosed that in the PML pathway, loss of PPAR5 and accompanying inhibition of mitochondrial FAO induced loss of hematopoietic stem cell (HSC) maintenance (Nature Medicine 18, 1350-1358 (2012)). Moreover, Carracedo et ah, disclosed that whereas PML expression allowed luminal filling in 3D basement membrane breast cancer, the effect was reversed by inhibition of FAO (J. Clin. Invest. 2012; 122(9):3088-3100). This and other evidence support our view that inhibition of fatty acid oxidation, via antagonism of PPAR's (including PPARa), will prove effective in inhibiting leukemia stem cell differential, and therefore, prove effective in preventing the onset of and/or recurrence of acute and chronic myeloid leukemia, as well as other cancers.
[0007] PPARa antagonists have also been shown to inhibit HCV replication and thereby prove useful in the treatment of HCV infection (Rakic, B. et. ah, Chem. & Biol. 13, 23-30 (January 2006)). In some embodiments, PPAR modulators have been shown to inhibit viral transcription and replication and thereby prove useful in the treatment of viral diseases (Capeau et ah, PPAR Research Volume 2009, Article ID 393408, 2 pages). In some embodiments, PPARa antagonists are useful in the treatment of HIV infection. PPARa antagonists have also been disclosed to be useful in the treatment of metabolic disorders (WO2012/027482A2). Metabolic disorders include, but are not limited to diabetes, obesity, metabolic syndrome, impaired glucose tolerance, syndrome X, and cardiovascular disease.
SUMMARY OF THE INVENTION
[0008] The invention disclosed herein is directed to compounds of Formula I
Figure imgf000003_0001
and pharmaceutically acceptable salts thereof, which are useful in the treatment of prostate, breast, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma, and other cancers. The invention also includes pharmaceutical compositions comprising a therapeutically effective amount of compound of Formula I, or a pharmaceutically acceptable salt thereof. The invention disclosed herein is also directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers. The invention disclosed herein is further directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers through the administration of a therapeutically effective amount of a selective PPARa antagonist. The compounds and pharmaceutical compositions of the invention are also useful in the treatment of viral infections, such as HCV infections and HIV infections.
INCORPORATION BY REFERENCE
[0009] All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.
DETAILED DESCRIPTION OF THE INVENTION
[0010] In one aspect the invention is directed to compounds of Formula I
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently -C^alkyl, optionally mono-, di- or tri-substituted with halogen, or
R1 and R2 are joined together to form -C3.6cycloall<yl, optionally mono- or di-substituted with substituents independently selected from halogen, -C^alkyl and -CF3;
R3 is selected from the group consisting of:
Figure imgf000004_0002
(b) aryl,
(c) heteroaryl,
Figure imgf000004_0003
wherein the aryl of choice (b) and the heteroaryl of choice (c) are optionally mono- or di-substituted with substituents independently selected from -N(H)R8, -Ci_6alkyl,
Figure imgf000005_0001
-C|.6alkoxy, halo-C|.
Figure imgf000005_0002
R4 is selected from the group consisting of:
(a) aryl,
(b) heteroaryl,
(c) -Ci_2alkyl-aryl, and
(d) -Ci_2alkyl-heteroaryl,
wherein the aryl of choices (a) and (c), and the heteroaryl of choices (b) and (d), are optionally mono- or di- substituted with substituents selected from the group consisting of halogen, -Ci_6alkyl, -CF3, -OCF3 -Ci_ 6alkoxy, -C3.6cycloalkoxy, halo-C|.6alkyl, aryl, heteroaryl, heterocyclo, -C3.6cycloalkyl, and -C3.6cycloalkenyl;
R5 and R6 are each independently selected from the group consisting of:
(a) hydrogen,
(b) -C^aHcyl,
(c) aryl, and
(d) hetereoaryl,
wherein the aryl of choice (c) and heteroaryl of choice (d) are optionally mono- or di-substituted with substituents independently selected from halogen, -Ci_6alkyl and -CF3 or
R5 and R6 are joined together to form a -C3.6cycloalkyl optionally mono- or di-substituted with substituents independently selected from halogen,
Figure imgf000005_0003
and -CF3;
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3_6cycloalkyl,
(e) -N(H)S(=0)2-Cwalkyl,
(f) -N(H)-aryl,
(g) -N(H)-heteroaryl,
(h) -N(H)-C3.6cycloalkyl,
(i) -N(H)-C1.6alkyl, and
(j) -CF3,
wherein the aryl of choices (b) and (f), the heteroaryl of choices (c) and (g), the alkyl portion of choices (e) and (i), and the cycloalkyl portion of choices (d) and (h), are optionally mono- or di-substituted with substituents independently selected from halogen, -Ci_6alkyl, -CF3, -C3.6cycloalkyl, -Ci_6alkoxy, -C3_
6cycloalkoxy, aryl, heteroaryl, and hydroxyl; and R is selected from the group consisting of:
(a) hydrogen,
(b) -S(=0)2aryl,
(c) -S(=0)2heteroaryl,
(d) -C(=0)aryl,
(e) -C(=0)heteroaryl,
(f) -S(=0)2-C1.6alkyl,
(g) -S(=0)2-C3-6cyc1oa1kyl,
(h) -C(=0)-C,.6alkyl, and
(i) -C(=0)-C3-6cycloalkyl,
wherein the aryl of choices (b) and (d), and the heteroaryl of choices (c) and (e), the alkyl portion of choices (f) and (h), and the cycloalkyl portion of choices (g) and (i), are optionally mono- or di-substituted with substituents independently selected from halogen, -Ci_6alkyl, -CF3, -C3_6cycloalkyl, -d-6alkoxy, -C3.
6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
[001 11 Within this aspect there is a genus wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen or
R1 and R2 are joined together to form -C3-6cycloalkyl, optionally mono or di-substituted with substituents independently selected from halogen, -Ci_6alkyl and -CF3.
[0012] Within this genus there is a sub-genus wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen.
[0013] Within this aspect there is a genus of Formula la
Figure imgf000006_0001
la
or a pharmaceutically acceptable salt thereof.
[0014] Within this aspect there is a genus wherein:
R4 is selected from the group consisting of:
(a) -phenyl,
(b) -pyridyl,
(c) -CH2-phenyl, and
(d) -CH2-pyridyl,
wherein the phenyl portion of choices (a) and (c), and the pyridyl portion of choices (b) and (d), are optionally mono- or di-substituted with substituents selected from the group consisting of halogen, -C|.6alkyl, -CF3, - OCF3, -Ci-6alkoxy, -C3-6cycloalkyl , -C3.(,cycloalkoxy, and halo-Ci_6alkyl.
[0015] Within this aspect there is a genus wherein: R5 and R6 are joined together to form a -C .6cycloalkyl.
[0016] Within this aspect there is a genus wherein:
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3-6cycloalkyl, and
(e) -N(H)S(=0)2-C,_6alkyl,
wherein the aryl portion of choice (b), the heteroaryl portion of choice (c), the cycloalkyl portion of choice (d) and the alkyl portion of choice (e) are optionally mono- or di-substituted with substituents independently selected from halogen, -C^alkyl, -CF3, -C3_6cycloalkyl, -C^alkoxy, -C3_6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
[0017] Within this genus there is a sub-genus wherein:
R7 is hydroxyl.
[0018] Within this aspect there is a genus wherein:
Rs is selected from the group consisting of:
(a) hydrogen,
(b) -S(=0)2aryl,
(c) -S(=0)2heteroaryl,
(d) -C(=0)aryl, and
(e) -C(=0)heteroaryl,
wherein the aryl of choices (b) and (d), and the heteroaryl of choices (c) and (e), are optionally mono- or di- substituted with substituents independently selected from halogen, -C^alkyl, -CF3, -C3.6cycloalkyl, -Ci_ 6alkoxy, -C3.6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
[0019] Within this aspect there is a genus of Formula la
Figure imgf000008_0001
la
or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen or
R1 and R2 are joined together to form -C3.6cycloalkyl, optionally mono or di-substituted with substituents independently selected from halogen, -d_6alkyl and -CF3;
R4 is selected from the group consisting of:
(a) -phenyl,
(b) -pyridyl,
(c) -CH2-phenyl, and
(d) -CH2-pyridyl,
wherein the phenyl portion of choices (a) and (c), and the pyridyl portion of choices (b) and (d), are optionally mono- or di-substituted with substituents selected from the group consisting of halogen, -Ci_6alkyl, -CF3, - OCF3, -Ci_6alkoxy, -C3.6cycloall<yl , -C3.6cycloalkoxy, and halo-Ci_6alkyl;
R5 and R6 are joined together to form a -C3.6cycloalkyl; and
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3.6cycloalkyl, and
(e) -N(H)S(=0)2-C,_6alkyl,
[0020] wherein the aryl portion of choice (b), the heteroaryl portion of choice (c), the cycloalkyl portion of choice (d) and the alkyl portion of choice (e) are optionally mono- or di-substituted with substituents independently selected from halogen, -C^alkyl, -CF3, -C3.6cycloalkyl, -C^alkoxy, -C3.6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
[0021] Within this genus there is a sub-genus wherein:
R7 is hydroxyl.
[0022] In another aspect the invention is directed to a pharmaceutical composition comprising a compound of Formula I (and/or a compound of any of the other formulae described herein), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
[0023] In another aspect the invention is directed to a method of treating a cancer which is negatively impacted by diminution in its metabolism of fatty acid oxidation via the adminstration of a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof. [0024] Within this aspect there is a genus wherein the cancer is selected from prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, and melanoma.
[0025] In another aspect the invention is directed to a method of treating cancer comprising the administration of a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
[0026] In another aspect the invention is directed to a method of preventing the onset of and/or recurrence of acute and chronic myeloid leukemia, as well as other cancers, through the administration of a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.
DEFINITIONS
[0027] The term "patient" includes mammals such as mice, rats, cows, sheep, pigs, rabbits, goats, horses, monkeys, dogs, cats, and humans.
[0028] The term "halo" or "halogen" refers to any radical of fluorine, chlorine, bromine or iodine.
[0029] The term "alkyl" refers to a saturated hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, Ci_6alkyl indicates that the group may have from 1 to 6 (inclusive) carbon atoms in it. In some embodiments, an alkyl is a C^alkyl. Any atom can be optionally substituted, e.g., by one or more substitutents. Examples of alkyl groups include without limitation methyl, ethyl, «-propyl, isopropyl, «-butyl, sec-butyl and tert-butyl.
[0030] The term "haloalkyl" refers to an alkyl group, in which at least one hydrogen atom is replaced by halo. In some embodiments, more than one hydrogen atom (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, or 14) are replaced by halo. In these embodiments, the hydrogen atoms can each be replaced by the same halogen (e.g., fluoro) or the hydrogen atoms can be replaced by a combination of different halogens (e.g., fluoro and chloro). "Haloalkyl" also includes alkyl moieties in which all hydrogens have been replaced by halo (sometimes referred to herein as perhaloalkyl, e.g., perfluoroalkyl, such as trifluoromethyl). Any atom can be optionally substituted, e.g., by one or more substituents. In some embodiments, a haloalkyl is a C^haloalkyl. In some embodiments, a fluoroalkyl is a Ci_6fluoroalkyl.
[0031] As referred to herein, the term "alkoxy" refers to a group of formula -O-(alkyl). Alkoxy can be, for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, wo-butoxy, sec-butoxy, pentoxy, 2-pentoxy, 3- pentoxy, or hexyloxy. Likewise, the term "thioalkoxy" refers to a group of formula -S-(alkyl). The terms "haloalkoxy" and "thioalkoxy" refer to -O-(haloalkyl) and -S-(haloalkyl), respectively. The term "sulfhydryl" refers to -SH.
[0032] The term "aralkyl" refers to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group. One of the carbons of the alkyl moiety serves as the point of attachment of the aralkyl group to another moiety. Any ring or chain atom can be optionally substituted e.g., by one or more substituents. Non-limiting examples of "aralkyl" include benzyl, 2-phenylethyl, and 3-phenylpropyl groups. [0033] The term "alkenyl" refers to a straight or branched hydrocarbon chain containing the indicated number of carbon atoms and having one or more carbon-carbon double bonds. Any atom can be optionally substituted, e.g., by one or more substituents. Alkenyl groups can include, e.g., vinyl, allyl, 1 -butenyl, and 2-hexenyl. In some embodiments, an alkenyl is a C2.6alkenyl.
[0034] The term "heterocycle" or "heterocyclic" includes heterocycloalkyls and heteroaryls.
[0035] The term "heterocycloalkyl" as used herein except where noted, represents a stable 3-, 4-, 5-, 6- or 7- membered monocyclic- or stable 6-, 7-, 8-, 9-, 10-, 1 1-, or 12-membered fused bicyclic heterocyclic ring system which comprises at least one non-aromatic (i.e. saturated or partially unsaturated) ring which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S, wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and wherein the nitrogen heteroatom may optionally be quaternized. In some embodiments, a heterocycloalkyl is a C2.ioheterocycloalkyl. In other embodiments, a heterocycloalkyl is a C2-6heterocycloalkyl. In some embodiments, a heterocycloalkyl is monocyclic. In some embodiments, a heterocycloalkyl is bicyclic. In the case of a "heterocycloalkyl" which is a bicyclic group, the second ring may also be a non-aromatic ring which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S, as defined above, or the second ring may be a benzene ring, or a "cycloalkyl", or a "cycloalkenyl", as defined immediately below. Examples of such heterocyclic groups include, but are not limited to, aziridine, azetidine, chroman, dihydrofuran, dihydropyran, dioxane, dioxolane, hexahydroazepine, imidazolidine, imidazoline, indoline, isochroman, isoindoline, isothiazoline, isothiazolidine, isoxazoline, isoxazolidine, morpholine, oxazoline, oxazolidine, oxetane, piperazine, piperidine, pyran, pyrazolidine, pyrazoline, pyrrolidine, pyrroline, tetrahydrofuran, tetrahydropyran, thiamorpholine, thiazoline, thiazolidine, thiomorpholine, and N-oxides thereof.
[0036] The term "cycloalkyl" refers to a fully saturated monocyclic, bicyclic, tricyclic, or other polycyclic hydrocarbon groups. Any atom can be optionally substituted, e.g., by one or more substituents. A ring carbon serves as the point of attachment of a cycloalkyl group to another moiety. Cycloalkyl moieties can include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, and norbornyl (bicycle[2.2.1]heptyl). In some embodiments, a cycloalkyl is a C3_iocycloalkyl. In other embodiments, a cycloalkyl is a C3.6cycloalkyl. In some embodiments, a cycloalkyl is monocyclic. In some embodiments, a cycloalkyl is bicyclic.
[0037] The term "cycloalkenyl" refers to partially unsaturated monocyclic, bicyclic, tricyclic, or other polycyclic hydrocarbon groups. A ring carbon (e.g., saturated or unsaturated) is the point of attachment of the cycloalkenyl substituent. Any atom can be optionally substituted e.g., by one or more substituents. Cycloalkenyl moieties can include, e.g., cyclopentenyl, cyclohexenyl, cyclohexadienyl, or norbornenyl. In some embodiments, a cycloalkenyl is a C 0cycloalkenyl. In other embodiments, a cycloalkenyl is a C4_ 6cycloalkenyl. In some embodiments, a cycloalkenyl is monocyclic. In some embodiments, a cycloalkenyl is bicyclic.
[0038] The term "cycloalkylene", as used herein, refers to a divalent monocyclic cycloalkyl group having the indicated number of ring atoms. [0039] The term "heterocycloalkylene", as used herein, refers to a divalent monocyclic heterocyclyl group having the indicated number of ring atoms.
[0040] The term "aryl" as used herein, is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, or biphenyl.
[0041] The term "heteroaryl", as used herein except where noted, represents a stable 5-, 6- or 7-membered monocyclic- or stable 9 or 10-membered fused bicyclic ring system which comprises at least one aromatic ring, -which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. In the case of a "heteroaryl" which is a bicyclic group, the second ring need not be aromatic and need not comprise a heteroatom. Accordingly, "heteroaryl" includes, for example, a stable 5-, 6- or 7-membered monocyclic aromatic ring consisting of carbon atoms and from one to four heteroatoms, as defined immediately above, fused to a benzene ring, or fused to a "heterocycloalkyl", a "cycloalkyl", or a "cycloalkenyl", as defined above. Examples of such heteroaryl groups include, but are not limited to, benzimidazole, benzisothiazole, benzisoxazole, benzofuran, isobenzofuran, benzothiazole, benzothiophene, benzotriazole, benzoxazole, carboline, cinnoline, furan, furazan, imidazole, indazole, indole, indolizine, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, phthalazine, pteridine, purine, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinazoline, quinoline, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazine, triazole, and N-oxides thereof.
[0042] The term "acyl", as used herein, refers to those groups derived from an organic acid by removal of the hydroxy portion of the acid. Accordingly, acyl is meant to include, for example, acetyl, propionyl, butyryl, decanoyl, pivaloyl, benzoyl and the like.
COMPOUND FORMS AND SALTS
[0043] The compounds of this invention may contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, enantiomerically enriched mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. The compounds of this invention include all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. The compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein, even though only a single tautomeric form may be represented. All such isomeric forms of such compounds are expressly included in the present invention.
[0044] The compounds of this invention include the compounds themselves, as well as their salts and their prodrugs, if applicable. A salt, for example, can be formed between an anion and a positively charged substituent (e.g., amino) on a compound described herein. Suitable anions include chloride, bromide, iodide, sulfate, nitrate, phosphate, citrate, methanesulfonate, trifluoroacetate, and acetate. Likewise, a salt can also be formed between a cation and a negatively charged substituent (e.g., carboxylate) on a compound described herein. Suitable cations include sodium ion, potassium ion, magnesium ion, calcium ion, and an ammonium cation such as tetramethylammonium ion.
[0045] As used herein, "pharmaceutically acceptable salts" refer to derivatives wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
[0046] When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, jo-toluenesulfonic acid, and the like. In one aspect of the invention the salts are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids.
[0047] When the compound of the present invention is acidic, salts may be prepared from pharmaceutically acceptable non-toxic bases, including inorganic and organic acids. Such salts that may be prepared include lithium salt, sodium salt, potassium salt, magnesium salt, calcium salt, dicyclohexylamine salt, N-methyl-D- glucamine salt, tris(hydroxymethyl)methylamine salt, arginine salt, lysine salt, and the like.
[0048] Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418; Journal of Pharmaceutical Science, 66, 2 (1977); and "Pharmaceutical Salts: Properties, Selection, and Use. A Handbook"; Wermuth, C. G. and Stahl, P. H. (eds.) Verlag Helvetica Chimica Acta, Zurich, 2002 [ISBN 3-906390-26-8] each of which is incorporated herein by reference in their entireties.
[0049] The compounds may be radiolabeled with radioactive isotopes, such as for example tritium, iodine- 125 or carbon-14. All isotopic variations of the compounds of the invention, whether radioactive or not, are intended to be encompassed within the scope of the invention.
[0050] In some embodiments, hydrogen atoms of the compounds described herein may be replaced with deuterium atoms.
[0051] In some embodiments, compounds of Formula I are prepared as prodrugs. Prodrugs are generally drug precursors that, following administration to a subject and subsequent absorption, are converted to an active, or a more active species via some process, such as conversion by a metabolic pathway. Examples of prodrugs include Ci_6 alkyl esters of carboxylic acid groups, which, upon administration to a subject, are capable of providing active compounds.
PHARMACEUTICAL COMPOSITIONS
[0052] The term "pharmaceutically acceptable carrier" refers to a carrier or an adjuvant that may be administered to a patient, together with a compound of this invention, or a pharmaceutically acceptable salt thereof, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
[0053] The term "composition" as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term in relation to pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
[0054] The present invention includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the terms "administration of or "administering a" compound shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs," ed. H. Bundgaard, Elsevier, 1985. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.
[0055] The amount administered depends on the compound formulation, route of administration, etc. and is generally empirically determined in routine trials, and variations will necessarily occur depending on the target, the host, and the route of administration, etc. Generally, the quantity of active compound in a unit dose of preparation may be varied or adjusted from about 1 , 3, 10 or 30 to about 30, 100, 300 or 1000 mg, according to the particular application. For convenience, the total daily dosage may be divided and administered in portions during the day if desired.
USES [0056] In one aspect the invention disclosed herein is directed to compounds of Formula I and pharmaceutically acceptable salts thereof, which are useful in the treatment of prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers. In another aspect, the invention is directed to a method of preventing the onset of and/or recurrence of acute and chronic myeloid leukemia, as well as other cancers. The invention also includes pharmaceutical compositions comprising a therapeutically effective amount of compound of Formula I, or a pharmaceutically acceptable salt thereof. The invention disclosed herein is also directed to methods of treating prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers. The invention disclosed herein is further directed to methods of treating prostate, breast, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers comprising administration of a therapeutically effective amount of a selective PPARa antagonist. The methods include administering to the subject an effective amount of a compound of Formula (I) (and/or a compound of any of the other formulae described herein) or a salt (e.g., a pharmaceutically acceptable salt) thereof as defined anywhere herein to the patient. In another aspect, the use of a compound of Formula (I) (and/or a compound of any of the other formulae described herein) or a salt (e.g., a pharmaceutically acceptable salt) thereof as defined anywhere herein in the preparation of, or for use as, a medicament for the treatment (e.g., controlling, alleviating, or slowing the progression of) or prevention (e.g., delaying the onset of or reducing the risk of developing) of one or more diseases, disorders, or conditions caused by, or associated with, prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, acute or chronic myeloid leukemia, melanoma and other cancers.
[0057] In one aspect the invention is directed a method of treating a cancer which is negatively impacted by diminution in its metabolism via fatty acid oxidation, comprising administration of a therapeutically effective amount of a compound of Formula I (and/or a compound of any of the other formulae described herein) or a salt (e.g., a pharmaceutically acceptable salt) there of. In another aspect, the invention is directed to a method of treating a cancer having a metabolism that is reliant on fatty acid oxidation, comprising administration of a therapeutically effective amount of a compound of Formula I (and/or a compound of any of the other formulae described herein), or a pharmaceutically acceptable salt thereof.
ADMINISTRATION
[0058] The compounds and compositions described herein can, for example, be administered orally, parenterally (e.g., subcutaneously, intracutaneously, intravenously, intramuscularly, intraarticularly, intraarterially, intrasynovially, intrasternally, intrathecally, intralesionally and by intracranial injection or infusion techniques), by inhalation spray, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, by injection, subdermally, intraperitoneally, transmucosally, or in an ophthalmic preparation, with a dosage ranging from about 0.01 mg/kg to about 1000 mg/kg, (e.g., from about 0.01 to about 100 mg/kg, from about 0.1 to about 100 mg/kg, from about 1 to about 100 mg/kg, from about 1 to about 10 mg/kg) every 4 to 120 hours, or according to the requirements of the particular drug. The interrelationship of dosages for animals and humans (based on milligrams per meter squared of body surface) is described by Freireich et al., Cancer Chemother. Rep. 50, 219 (1966). Body surface area may be approximately determined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardsley, N.Y., 537 (1970). In certain embodiments, the compositions are administered by oral administration or by injection. The methods herein contemplate administration of an effective amount of compound or compound composition to achieve the desired or stated effect. Typically, the pharmaceutical compositions of this invention will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
[0059] Lower or higher doses than those recited above may be required. Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, and the judgment of the treating physician.
[0060] Dosage forms include from about 0.05 milligrams to about 2,000 milligrams (e.g., from about 0.1 milligrams to about 1 ,000 milligrams, from about 0.1 milligrams to about 500 milligrams, from about 0.1 milligrams to about 250 milligrams, from about 0.1 milligrams to about 100 milligrams, from about 0.1 milligrams to about 50 milligrams, or from about 0.1 milligrams to about 25 milligrams) of a compound of Formula I (and/or a compound of any of the other formulae described herein) or a salt (e.g., a pharmaceutically acceptable salt) thereof as defined anywhere herein. The dosage forms can further include a pharmaceutically acceptable carrier and/or an additional therapeutic agent.
[0061] In one aspect the compounds of the invention may be co-administered with one or more additional anti-cancer agents. The additional anti-cancer agents include, but are not limited to alkylating agents such as cyclophosphamide, chlorambucil, mecloreethamine, ifosfamide, or melphalan; antimetabolites such as methotrexate, cytarabine, fludarabine, 6-mercaptopurine, azathioprene, pyrimidines, or 5-fluorouracil; antimitotic agents such as vincristine, paclitaxel, vinorelbine or docetaxaxel; a topoisomerase inhibitors such as doxorubicin or irinotecan; platinum derivatives such as cisplatin, carboplatin or oxaliplatin; hormone therapeutics such as tamoxifen; aromatase inhibitors such as bicalutamide, anastrozole, exemestane or letrozole; signaling inhibitors such as imatinib, gefitinib or erlotinib; monoclonal antibodies such as rituximab, trastuzumab, gemtuzumab or ozogamicin; differentiating agents such as tretinoin or arsenic trioxide; antiangiogenic agents such as bevacizumab, sorafinib or sunitinib; biologic response modifiers such as interferon-alpha; topoisomerase inhibitors such as camptothecins (including irinotecan and topotecan), amsacrine, etoposide, etoposide phosphate, or teniposide; cytotoxic antibiotics such as actinomycin, anthracyclines including doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin or mitomycin; vinca alkaloids such as vincristine, vinblastine, viorelbine or vindesine; podophyllotoxins such as etoposide and teniposide; or mTOR inhibitors such as rapamycin, temsirolimus and everolimus. [0062] Other anti-cancer agents for use in combination with the compounds include one or more of the following: abiraterone; adriamycin; acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; everolimus; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; iimofosine; interleukin II (including recombinant interleukin II, or rlL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-nl ; interferon alfa- n3 ; interferon beta-1 a; interferon gamma-1 b; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazoie; nogalamycin; ormaplatin; oxisuran; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; rapamycin; riboprine; rogletimide; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; temsirolimus; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride.
[0063] In certain embodiments, the additional agents may be administered separately (e.g., sequentially; on different overlapping schedules), as part of a multiple dose regimen, from the compounds of this invention (e.g., one or more compounds of Formula (I) and/or a compound of any of the other formulae, including any subgenera or specific compounds thereof). In other embodiments, these agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition. In still another embodiment, these agents can be given as a separate dose that is administered at about the same time as that of one or more compounds of Formula (I) (and/or a compound of any of the other formulae, including any subgenera or specific compounds thereof) are administered (e.g., simultaneously with the administration of one or more compounds of Formula (I) (and/or a compound of any of the other formulae, including any subgenera or specific compounds thereof)). When the compositions of this invention include a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents, both the compound and the additional agent can be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
[0064] The compositions of this invention may contain any conventional non-toxic pharmaceutically- acceptable carriers, adjuvants or vehicles. In some cases, the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
[0065] The compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions and/or emulsions are administered orally, the active ingredient may be suspended or dissolved in an oily phase and then combined with emulsifying and/or suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
BIOLOGICAL FUNCTION
[0066] The utility of the invention can be demonstrated by one or more of the following methods or other methods known in the art:
Human PPARa Reporter Assay
[0067] The screening of test compounds for agonist or antagonist activities against human PPARa receptors was performed using a commercial kit, Human PPARa Reporter Assay System (Indigo Biosciences, Cat. #IB001 1 1 ).
[0068] This nuclear receptor assay system utilizes proprietary non-human mammalian cells engineered to provide constitutive, high-level expression of Human PPARa. Because these cells incorporate a PPARa- responsive luciferase reporter gene, quantifying expressed luciferase activity provides a sensitive surrogate measure of PPARa activity in the treated cells. The primary application of this reporter assay system is in the screening of test samples to quantify any functional activity, either agonist or antagonist, that they may exert against human PPARa.
[0069] While this assay may be used to measure agonism, each of the Examples, vide infra, exhibits antagonism rather than agonism. Briefly, reporter cells are dispensed into wells of the assay plate and then immediately dosed with test compounds. Following an overnight incubation, the treatment media is discarded and Luciferase Detection Reagent (LDR) is added. The intensity of light emission from the ensuing luciferase reaction provides a sensitive measure that is directly proportional to the relative level of PPARa activation in the reporter cells.
Figure imgf000018_0001
Figure imgf000019_0001
*mass spectroscopic data
Target Selectivity Assays
[0070] To determine species selectivity, a Mouse PPARa Reporter Assay System was used (Indigo Biosciences, Cat. #M001 1 1 ). Activity of test compounds to antagonize or agonize other isoforms of human PPAR, for example β/δ and γ, were assessed using the corresponding kits from Indigo Biosciences (Cat. #IB00121 and #IB00101 , respectively). In addition to PPAR activity, compounds were also screened for activity against other nuclear hormone receptors including Estrogen Receptor β, Glucocorticoid Receptor and Thyroid Receptor β using commercially available kits (Indigo Biosciences, Cat. #IB0041 1 , IB00201 and IBOl lO l, respectively). Each assay system from Indigo Biosciences uses technology analogous to the human PPARa kit, with the variance being that the cells used for each assay were engineered to over-express the receptor of interest. In addition, the appropriate receptor agonist (included with each kit) was used at ~EC80 for assays in which antagonist potency was being assessed.
Target Selectivity - Counterscreen Assay Results
Figure imgf000020_0001
Measurement of cell viability
[0071] Purified CLL cells were cultured at 2 x 10s cells/200 μΐ, of RPMI1640 supplemented with 10% FCS in 96-well plates under various treatment conditions. Determination of CLL cell viability was based on the analysis of mitochondrial transmembrane potential (ΔΨηι) using 3,3'-dihexyloxacarbocyanine iodide (DiOC6) (Invitrogen) and cell membrane permeability to propidium iodide (PI) (Sigma). For viability assays, 100 of the cell culture was collected at the indicated time points and transferred to polypropylene tubes containing 100 μΐ. of 40 μΜ DiOC6 and 10 μg/mL PI in culture media. The cells were then incubated at 37 °C for 15 min and analyzed within 30 min by flow cytometry using an Accuri C6 flow cytometer. The percentage of viable cells was determined by gating on PI negative and DiOC6 bright cells.
In vivo cancer model: B16F10 model of pulmonary metastasis
[0072] B 16F 10 cells are cultured in standard growth media, harvested when approximately 50% confluent and injected into C57BL/6 mice via the tail vein (50,000 cells per mouse in 200 μΕ). Mice are then treated daily with test compound. On day 21 , mice are euthanized. Lungs are harvested and placed into Fekete's solution overnight to facilitate visualization of the tumors. Black nodules are enumerated.
[0073] Statistics are performed by ANOVA with Dunnett's Multiple Comparison Test post-hoc to determine statistical differences from vehicle treatment group (* denotes PO.05 while *** denotes P<0.001 ).
SYNTHESIS
[0074] The starting materials used for the synthesis are either synthesized or obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Fluka, Acros Organics, Alfa Aesar, VWR Scientific, and the like. General methods for the preparation of compounds can be modified by the use of appropriate reagents and conditions for the introduction of the various moieties found in the structures as provided herein.
[0075] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby. General Synthetic Scheme for Exemplary Compounds
[0076] The pyrazalone core found in the exemplified compounds could be conveniently accessed, for example, from the corresponding carboxylic acid (I, Scheme 1) via its initial decarboxylative Claisen condensation with an appropriate monoester of malonic acid (II, Scheme 1). This condensation can, for example, be carried out in the presence of an appropriate activating reagent such as carbonyldiimidazole (i.e. CDI), an appropriate Lewis acid such as magnesium chloride, and an appropriate base such as triethylamine (i.e. TEA). The resulting β-ketoester III could then be di-alkylated, either sequentially or in one-pot, by, for example, its treatment with an appropriate base such as NaH, Cs2C03, K2C03, or others, followed by the addition of a suitable alkylating reagent, denoted generically as R'-X and R2-X in scheme 1. In examples where R1 and R2 are joined to form a ring, the requisite intermediate could most conveniently be synthesized using, for example, ring closing metathesis whereby the olefin functional group present in both R1 and R2 in IV are welded together by a suitable mediator, such as Grubb's (both generation 1 and generation 2) or Schrock's metathesis catalyst, with the concomitant extrusion of ethene gas. Subsequent hydrogenation of the resulting cyclic alkene, using for example hydrogen gas and palladium, would deliver the requisite saturated carbacyle V. Heating either IV or V with hydrazine in an alcohol solvent such as, for example, ethanol or methanol, in an enclosed reaction vessel would then furnish pyrazolone VI. This could in turn be functionalized, as appropriate, via either a simple nucleophilic displacement reaction (in instances where R4 is an alkyl or heteroalkyl group) or via copper catalyzed N-arylation reaction (in instances where R4 is an aryl or heteroaryl group). In the former case, a base such as, though not limited to, NaH, Cs2C03 or 2C03 is added along with R4-X to pyrazalone VI in a polar, aprotic solvent. In the latter case, a convenient source of copper(I) such as copper iodide, a suitable ligand such as proline, and a suitable base such as Cs2C03, are heated along with to R4-X to pyrazolone VI in a polar, aprotic solvent.
Scheme 1
Figure imgf000022_0001
[0077] Functionalization of pyrazalone VII can also be readily achieved (Scheme 2). In instances where Y is OH, O-alkylation can be conveniently accomplished by reacting VII with, for example, an activated halide such as VIII in the presence of an appropriate base such as Cs2C03, NaOEt, NaH, or the like. The resulting fibrate ester IX can then be hydrolyzed to deliver the corresponding acid X. The acid can be further coupled with an amine (to give amide XI) or a sulfonamide (to deliver acyl sulfonamide XII) via the use of an appropriate coupling reagent such as HATU, EEDQ, EDC, CDI, or the like, and in the presence of an appropriate base such as TEA, Hunig's base, DABCO, DMAP, or the like. Alternatively, acid X could be reduced to its corresponding alcohol XIV, by its initial conversion to the mixed anhydride XIII and subsequent treatment with agents such as sodium borohydride, lithium borohydride, or the like. In turn, primary alcohol XIV can be converted to the secondary alcohol XV via a two-step protocol: (1 ) oxidation with reagents such as Dess-Martin periodinane, S03-pyridine, Swern reagent, or the like, and (2) reaction with a suitable organometallic reagent depicted generically as R8-M. A subsequent oxidative event would deliver ketone XVI. Alternatively, alcohol XIV can be transformed to amine XVII using a variety of approaches known to those skilled in the art (e.g. the palladium-mediated reduction of an intermediate azide accessed using Curtius rearrangement). Further derivatization of amine XVII to amide XVIII (via, for example, acylation with R8-C(=0)C1) or to sulfonamide XIX (via, for example, sulfonylation with R8-S(=0)2C1) could also be carried out using standard procedures known to those skilled in the art. In instance were Y is CI, Br, or I (Scheme 2), metal -catalyzed cross-couplings such as the Suzuki reaction, the Stille reaction, the Negishi reaction, or the like, can be employed to facilitate further structural diversification from pyrazole VII. Once again, manipulations of the functional groups on biaryl XX (denoted generically as FG in Scheme 2) using some of, but not limited to, the chemical transformations described previously (i.e. acylation, sulfonylation, oxidation, reduction, alkylation, arylation, hydrolysis, addition or the like), can be readily envisioned.
Scheme 2
Figure imgf000024_0001
Example 1: 2-(4-(3-(l-(4-(fe^-butyl benzyl -4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3- vPpropyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000025_0001
[0078] Step 1: To an ethanol solution (0.8 M) of 4-(4-hydroxyphenyl)butanoic acid (1 eq.) was added freshly prepared sodium ethoxide (2 eq.). The resulting mixture was then heated at reflux for 30 min before ethyl 2- bromo-2-methylpropanoate (3 eq.) was added neat and dropwise over 5min. The now black reaction suspension was heated at reflux for another 2 h before another portion of sodium ethoxide (1 eq.) was added. The resulting mixture was then heated at reflux for 16 h before it was carefully quenched with the addition of 10% aq. HCl. After the removal of the volatiles in vacuo, the resulting brown oil was partitioned between EtOAc and water. The aqueous wash was separated and back-extracted with EtOAc. The combined organic extracts were washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the desired product as a tan solid (85% yield).
[0079] Step 2: To an acetonitrile solution (0.05 M) of 4-(4-((l -ethoxy-2-methyl-l -oxopropan-2- yl)oxy)phenyl)butanoic acid (1 eq.) from the previous step was added CDI (1.1 eq.). The resulting yellow solution was then allowed to stir at RT for 2.5 h before it was added dropwise, over a period of 1.5 h, into a white suspension of potassium 3-methoxy-3-oxopropanoate (2.1 eq.), magnesium chloride (2.5 eq.) and triethylamine (3.2 eq.). The resulting suspension was then stirred at RT for 16 h and finally heated at reflux for another 24 h. The crude reaction suspension thus obtained was cooled to RT and diluted with EtOAc. The insolubles were then removed via filtration and rinsed further with EtOAc and DCM. The filtrate thus obtained was concentrated in vacuo, re-taken up in EtOAc and washed sequentially with 10% aq. HCl, water and brine. The organic extract was then dried over Na2S04 and filtered. Concentration of the filtrate in vacuo affored the desired product as a golden oil (96% yield) which can be used without further purification.
[0080] Step 3: To a DMSO solution (0.1 M) of methyl 6-(4-((l -ethoxy-2-methyl-l -oxopropan-2- yl)oxy)phenyl)-3-oxohexanoate (1 eq.) from the previous step was added cesium carbonate (3 eq.) and iodomethane (3 eq.). The resulting mixture was then allowed to stir at RT for 16 h. The crude reaction mixture thus obtained was diluted with ether and washed sequentially with cold water, 10% aq. HCl, 1 N aq. NaOH, water and finally brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (74% yield).
[0081] Step 4: To an ethanol solution (0.7 M) of methyl 6-(4-((l-ethoxy-2 -methyl- 1 -oxopropan-2- yl)oxy)phenyl)-2,2-dimethyl-3-oxohexanoate (1 eq.) from the previous step was added hydrazine monohydrate (1.5 eq.). The reaction vessel was then tightly sealed and heated at 80 °C behind a blast shield for 48 h. After cooling to RT, the volatiles were then removed in vacuo and the resulting residue was partitioned between ether and 10% aq. HC1. The aqueous layer was separated and back-extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04 and filtered. Concentration fo the filtrate in vacuo afforded the desired product as a colorless oil (82% yield).
[0082] Step 5: To an acetonitrile solution (0.15 M) of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l//- pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added A-tert- butylbenzyl bromide (1 .2 eq.) and cesium carbonate (3 eq.). The resulting mixture was then heated at 60 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -^ 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (85% yield).
[0083] Step 6: To a 2: 1 (v/v) THF: MeOH solution (0.13 M) of ethyl 2-(4-(3-(l-(4-(teri-butyl)benzyl)-4,4- dimethyl-5-oxo-4,5-dihydro-l i/-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HC1 to pH of ~3. The aqueous suspension thus obtained was then extracted with EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the title compound as a white solid (96% yield). LC-MS: 479 (M+H)+.
Example 2: 2-(4-(3-(l-(4-(fe^-butyl benzyl -4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3- vnpropyl)phenoxy)-2-methyl-/V-(phenylsulfonyl)propanamide:
Figure imgf000026_0001
[0084] To a dichloromethane solution (0.1 M) of 2-(4-(3-( l -(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5- dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid (1 eq., Example 1) was added phenyl sulfonamide (1.4 eq.) and DMAP (1 eq.). The resulting mixture was sonicated briefly (~ 5 min) before EDC (1.4 eq.) was added neat and in one rapid portion. After 72 h, the volatiles were removed in vacuo and the resulting residue thus obtained was directly subjected to purification by way of reverse phase column chromatography (C18, gradient elution, 9: 1 (v/v) H20: MeCN + 0.1% TFA -> MeCN + 0.1% TFA) to furnish the title product as a white solid (42% yield). LC-MS: 618 (M+H)+. Exam pie 3 : jV-benzyl-2-(4-(3-(l -(4-(ferf-butyl)benzyl)-4,4-dim eth yl-5-oxo-4,5-dihyd ro- 1 //-pyrazol-3- yl)propyl)phenoxy -2-methylpropanamide:
Figure imgf000027_0001
[0085] To a DMF solution (0.1 M) of 2-(4-(3-(l-(4-(feri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//- pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid (1 eq., Example 1) was added benzylamine (1 eq.) and HATU (1.2 eq.). The resulting solution was sonicated briefly (~ 5 min) before Hunig's base (1.5 eq.) was added neat and in one rapid portion. After 5 h, the reaction mixture was diluted with ether and washed sequentially with water, 10% aq. HCl, 1 N aq. NaOH and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -^ 3 :7 (v/v) Hex: EtOAc) afforded the title compound as a colorless oil (88% yield). LC-MS: 568 (M+H)+.
Example 4: 2-(4-(3-(l-(4-(fe^-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3- vnpropyl)phenoxy)-2-methyl-jV-(pyridin-2-ylmethyl)propanamide:
Figure imgf000027_0002
[0086] Prepared in an analogous manner to Example 3 but using pyridin-2-ylmethanamine in place of benzylamine as the amine coupling partner. LC-MS: 569 (M+H)+.
Example 5: 2-(4-(3-(l-(4-(fe^-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3- yl)propyl)phenoxy -2-methyl-N-(pyridin-4-ylmethyl propanamide:
Figure imgf000028_0001
[0087] Prepared in an analogous manner to Example 3 but using pyridin-4-ylmethanamine in place of benzylamine as the amine coupling partner. LC-MS: 569 (M+H)+.
Example 6: 2-(4-(3-(l-(4-(fe^-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihvdro-l -pyrazol-3- v0propyl)phenoxy)-A^cvclohexylmethylV2-methylpropanamide:
Figure imgf000028_0002
[0088] Prepared in an analogous manner to Example 3 but using cyclohexylamine in place of benzylamine as the amine coupling partner. LC-MS: 574 (M+H)+.
Example 7: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(3-(trifluoromethyl)benzyl)-4,5-dihydro-l -pyrazol-3-
Figure imgf000028_0003
[0089] Prepared in an analogous manner to Example 1 but using 3-(trifluoromethyl)benzyl bromide in place of 4-teri-butylbenzyl bromide as the electrophile in step 5. LC-MS: 491 (M+H)+.
Example 8: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl benzyl -4,5-dihydro-l -pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000029_0001
[0090] Prepared in an analogous manner to Example 1 but using 4-(triiluoromethyl)benzyl bromide in place of 4-teri-butylbenzyl bromide as the electrophile in step 5. LC-MS: 491 (M+H)+.
Example 9: 2-(4-(3-(4,4-dimethyl-5-oxo-l-phenethyl-4,5-dihvdro-l -pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid:
Figure imgf000029_0002
[0091] Prepared in an analogous manner to Example 1 but using 2.8 eq. of (2-bromoethyl)benzene in place of 4-teri-butylbenzyl bromide as the electrophile in step 5. LC-MS: 437 (M+H)+.
Example 10: 2-(4-(3-(4,4-dimethyl-5-oxo-l-phenyl-4,5-dihvdro-l -pyrazol-3-yl)propynphenoxy)-2- methylpropanoic acid:
Figure imgf000029_0003
[0092] Step 1: To a DMSO solution (0.13 M) of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-lii-pyrazol- 3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq., Example 1 , Step 4) was added iodobenzene (1.5 eq.), copper(I) iodide (0.1 eq.), L-proline (0.2 eq.) and potassium carbonate (2.5 eq.). The resulting suspension was then deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. Then, the reaction vessel was tightly sealed and the reaction mixture was heated at 95 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil that solidified upon standing (76% yield).
[0093] Step 2: To a 2: 1 (v/v) THF: MeOH solution (0.05 M) of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-l -phenyl- 4,5-dihydro-l -pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HC1 to pH of ~3. The aqueous suspension thus obtained was then extracted with ether and EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the title compound as a white solid (99% yield). LC-MS: 409 (M+H)+.
Example 11: 2-(4-(3-(l-(4-(ferf-butyl)phenyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l y-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000030_0001
[0094] Prepared in an analogous manner to Example 10 but using l -(ierf-butyl)-4-iodobenzene in place of iodobenzene as the aryl iodide coupling partner in step 1. LC-MS: 465 (M+H)+.
Example 12: 2-(4-(3-(l-(4-isopropylbenzyl)-4,4-dimethyl-5-oxo-4,5-dihvdro-l//-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000030_0002
[0095] Prepared in an analogous manner to Example 1 but using l-(bromomethyl)-4-isopropylbenzene in place of 4-teri-butylbenzyl bromide as the electrophile in step 5. LC-MS: 465 (M+H)+.
Example 13: 2-(4-(3-(l-(4-bromobenzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l -pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000031_0001
[0096] Step 1: To an acetonitrile solution (0.07 M) of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l//- pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq., Example 1, Step 4) was added 4-bromobenzyl bromide (1 .2 eq.) and cesium carbonate (3 eq.). The resulting mixture was then heated at 60 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HCl and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (82% yield).
[0097] Step 2: To a 2: 1 (v/v) THF: MeOH solution (0.05 M) of ethyl 2-(4-(3-(l-(4-bromobenzyl)-4,4- dimethyl-5-oxo-4,5-dihydro-l i/-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HCl to pH of ~3. The aqueous suspension thus obtained was then extracted with ether and EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the title compound as a white solid (99% yield). LC-MS: 501. 503 (M+H)+.
Example 14: 2-(4-(3-(l-([lJ'-biphenyl1-4-ylmethyl)-4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3-
Figure imgf000031_0002
[0098] Step 1: To a 3 : 1 (v/v) DME: water solution (0.03 M) of ethyl 2-(4-(3-(l -(4-bromobenzyl)-4,4- dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq., Example 13, Step 1) was added phenylboronic acid (1 .2 eq.), tetrakis(triphenylphosphine)palladium(0) (0.1 eq.) and potassium carbonate (3 eq.). The resulting biphasic mixture was deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. Then, the reaction vessel was tightly sealed and the reaction mixture was heated at 90 °C for 16 h.. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -^ 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil (61% yield).
[0099] Step 2: To a 2: 1 (v/v) THF: MeOH solution (0.05 M) of ethyl 2-(4-(3-(l-([l, l 'biphenyl]-4-ylmethyl)- 4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HC1 to pH of ~3. The aqueous suspension thus obtained was then extracted with ether and EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the title compound as a white solid (95% yield). LC-MS: 499 (M+H)+.
Example 15: 2-(4-(3-(4,4-dimethyl-5-oxo-l-((2%3,,4%5,-tetrahvdro-| l ,l ,-biphenyl|-4-yl)methyl)-4,5-
Figure imgf000032_0001
[00100] Prepared in an analogous manner to Example 14 but using 1-cyclohexen-l -ylboronic acid, pinacol ester in place of phenylboronic acid as the coupling partner in step 1. LC-MS: 503 (M+H)+.
Figure imgf000033_0001
[00101] Prepared in an analogous manner to Example 14 but using palladium(II) acetate (0.1 eq.) and tricyclohexylphosphine (0.2 eq.) in place of tetrakis(triphenylphosphine)palladium(0) as the catalyst system, cyclopropylboronic acid (5 eq.) in place of phenylboronic acid as the coupling partner, potassium phosphate (3.5 eq.) in place of potassium carbonate as the base, and toluene in place of DME as the reaction co-solvent in step 1. LC-MS: 463 (M+H)+.
Example 17: 2-(4-(3-(l-(4-cvclohexylbenzyl)-4,4-dimethyl-5-oxo-4,5-dihvdro-l -pyrazol-3-
Figure imgf000033_0002
[00102] To an ethanol solution (0.01 M) of 2-(4-(3-(4,4-dimethyl-5-oxo-l -((2',3 ',4',5'-tetrahydro-[l , l '- biphenyl]-4-yl)methyl)-4,5-dihydro-l i-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid (1 eq., Example 15) was added palladium black (0.1 eq., 10% w/w (dry) over carbon). The resulting suspension was evacuated and back-filled with hydrogen gas (50 psi, 3x). Then, the reaction mixture was shaken on a Parr hydrogenator under 50 psi of hydrogen for 16 h. The mixture was carefully quenched with DCM and filtered through a bed of DCM-wetted celite. The insolubles were then washed further with DCM and EtOAc. Concentration of the filtrate thus obtained in vacuo furnished the title compound as a white solid (70% yield). LC-MS: 505 (M+H)+. Example 18: 2-(4-(3-(l-(3-fluoro-4-(trifluoromethyl benzvn-4,4-dimethyl-5-oxo-4,5-dihydro-l - Pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000034_0001
[00103] Prepared in an analogous manner to Example 1 but using 3-fluoro-4-(trifluoromethyl)benzyl bromide in place of 4-ierf-butylbenzyl bromide as the electrophile in step 5. LC-MS: 509 (M+H)+.
Example 19: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl phenyl)-4,5-dihvdro-l y-pyrazol-3-
Figure imgf000034_0002
[00104] Prepared in an analogous manner to Example 10 but using 4-iodobenzotrifluoride in place of iodobenzene as the aryl iodide coupling partner in step 1. LC-MS: 477 (M+H)+.
Example 20: 2-(4-(3-(4.,4-dimethyl-5-oxo-l-(3-(trifluoromethyl)phenyl)-4,5-dihvdro-l -pyrazol-3- vPpropyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000034_0003
[00105] Prepared in an analogous manner to Example 10 but using 3-iodobenzotrifluoride in place of iodobenzene as the aryl iodide coupling partner in step 1. LC-MS: 477 (M+H)+. Example 21: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(5-(trifluoromethyl pyridin-2-ylV4,5-dihvdro-lj -pyrazol-3- vPpropyl)phenoxy)-2-methylpropanoic acid:
Figure imgf000035_0001
[00106] Prepared in an analogous manner to Example 10 but using 2-iodo-5-(triiluoromethyl)pyridine in place of iodobenzene as the aryl iodide coupling partner in step 1. LC-MS: 478 (M+H)+.
Example 22: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l -pyrazol-3- vnpropyl)phenoxy)-2-methyl-jV-(phenylsulfonyl)propanamide:
Figure imgf000035_0002
[00107] Prepared in an analogous manner to Example 2 but using 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4- (trifluoromethyl)phenyl)-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid (Example 19) in place of 2-(4-(3-(l -(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid as the coupling partner. LC-MS: 616 (M+H)+.
Example 23: 2-(4-(3-(3-(4-(½rf-butyl)benzyl)-4-oxo-2,3-diazaspiro|4,4|non-l-en-l-yl)propyl)phenoxy)-2- methylpropanoic acid:
Figure imgf000035_0003
[00108] ¾ep 1: To a DMSO solution (0.1 M) of methyl 6-(4-((l-ethoxy-2-methyl-l -oxopropan-2- yl)oxy)phenyl)-3-oxohexanoate (1 eq., Example 1 , Step 2) was added cesium carbonate (3 eq.) and allyl bromide (3 eq.). The resulting mixture was then allowed to stir at RT for 16 h. The crude reaction mixture thus obtained was diluted with ether and washed sequentially with cold water, 10% aq. HCl, 1 N aq. NaOH, water and finally brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (62% yield).
[00109] Step 2: To a freshly deoxygenated 1 ,2-dichloroethane solution (0.002 M) of methyl 2,2-diallyl-6-(4- ((l -ethoxy-2 -methyl- l -oxopropan-2-yl)oxy)phenyl)-3-oxohexanoate (1 eq.) from the previous step was added Grubb's catalyst, second generation (0.05 eq.). The resulting mixture was then allowed to stir at RT, with a vent to allow for the outgassing of ethylene produced, for 16 h. The volatiles were then removed in vacuo and the resulting green residue was directly subjected to purification by way of column chromatography (Si02, gradient elution, 9: 1 -> 1 : 1 (v/v) Hex: EtOAc) to furnish the desired product as a colorless oil (89% yield).
[OOl l OJ Siep 3: To an ethyl acetate solution (0.07 M) of methyl l -(4-(4-((l -ethoxy-2-methyl-l -oxopropan-2- yl)oxy)phenyl)butanoyl)cyclopent-3-enecarboxylate (1 eq.) from the previous step was added palladium black (0.1 eq., 10% w/w (wet) over carbon). The resulting suspension was evacuated and back-filled with hydrogen gas (3x). Then, the reaction mixture was stirred under a static hydrogen atmosphere maintained with a balloon for 16 h. The mixture was carefully quenched with DCM and filtered through a bed of DCM-wetted celite. The insolubles were then washed further with DCM. Concentration of the filtrate thus obtained in vacuo furnished the desired compound as a colorless oil (94% yield).
[00111] Step 4: To an ethanol solution (0.18 M) of methyl l-(4-(4-((l-ethoxy-2 -methyl- 1 -oxopropan-2- yl)oxy)phenyl)butanoyl)cyclopentanecarboxylate (1 eq.) from the previous step was added hydrazine monohydrate (1 .5 eq.). The reaction vessel was then tightly sealed and heated at 80 °C behind a blast shield for 96 h. After cooling to RT, the volatiles were then removed in vacuo and the resulting residue was partitioned between ether and 10% aq. HCl. The aqueous layer was separated and back-extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -^ 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil that solidified upon standing (70% yield).
[00112] Step 5: To an acetonitrile solution (0.16 M) of ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non- l-en-l-yl)propyl)phenoxy)propanoate (1 eq.) from the previous step was added 4-ieri-butylbenzyl bromide (1.2 eq.) and cesium carbonate (3 eq.). The resulting mixture was then heated at 60 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HCl and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil (96% yield).
[00113] Step 6: To a 2: 1 (v/v) THF: MeOH solution (0.15 M) of ethyl 2-(4-(3-(3-(4-(ieri-butyl)benzyl-4-oxo- 2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)-2-methylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HCl to pH of ~3. The aqueous suspension thus obtained was then extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Trituration of the crude product thus obtained in hexanes afforded the title compound as a white solid (85% yield). LC-MS: 505 (M+H)+.
Example 24: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenylV2,3-diazaspiro|4,4|non-l-en-l- yl)propyl)phenoxy)propanoic acid:
Figure imgf000037_0001
[00114] Prepared in an analogous manner to Example 10 but using 4-iodobenzotrifluoride in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l ii-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 503 (M+H)+.
Example 25: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenylV2,3-diazaspiro|4,4|non-l-en-l-
Figure imgf000037_0002
[00115] Prepared in an analogous manner to Example 2 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l-yl)propyl)phenoxy)propanoic acid (Example 24) in place of 2-(4-(3-(l -(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid as the coupling partner. LC-MS: 642 (M+H)+.
Example 26: N-cvclopropyl-2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3- diazaspiro|4,4|non-l-en-l-yl)propyl)phenoxy)propanamide:
Figure imgf000037_0003
[00116] Prepared in an analogous manner to Example 3 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid (Example 24) in place of 2-(4-(3-(l -(4-(iert-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid, and cyclopropylamine in place of benzyl amine as the coupling partners. LC-MS: 542 (M+H)+.
Example 27: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl phenylV2,3-diazaspiro[4,41non-l-en-l- v0propyl)phenoxy)-A^2,2,2-trifluoroethy )propanamide:
Figure imgf000038_0001
[00117] Prepared in an analogous manner to Example 3 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid (Example 24) in place of 2-(4-(3-(l -(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid, and 2,2,2-trifluoroethylamine in place of benzyl amine as the coupling partners. LC- MS: 584 (M+H)+.
Example 28: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,41non-l-en-l- vPpropyl)phenoxy)propanoic acid:
Figure imgf000039_0001
[00118] Prepared in an analogous manner to Example 10 but using l -iodo-4-(trifluoromethoxy)benzene in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5- dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 519 (M+H)+.
Example 29: jV-cvclopropyl-2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3- diazaspiro|4,4|non-l-en-l-yl)propyl)phenoxy)propanamide:
Figure imgf000039_0002
[00119] Prepared in an analogous manner to Example 3 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l-en-l-yl)propyl)phenoxy)propanoic acid (Example 28) in place of 2-(4-(3-(l -(4-(iert-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid, and cyclopropylamine in place of benzyl amine as the coupling partners. LC-MS: 558 (M+H)+.
Example 30: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l-en-l- yl)propyl)phenoxy)-N-(2,2,2-trifluoroethyl)propanamide:
Figure imgf000039_0003
[00120] Prepared in an analogous manner to Example 3 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid (Example 28) in place of 2-(4-(3-(l -(4-(iert-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid, and 2,2,2-trifluoroethylamine in place of benzyl amine as the coupling partners. LC- MS: 600 (M+H)+.
Example 31: 2-(4-(3-(3-(3,4-dimethylphenyl -4-oxo-2,3-diazaspiro[4,41non-l-en-l-yl)propyl phenoxy)-2- methylpropanoic acid:
Figure imgf000040_0001
[00121] Prepared in an analogous manner to Example 10 but using 4-iodo-l ,2-dimethylbenzene in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-li7-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 463 (M+H)+.
Example 32: N-cvclopropyl-2-(4-(3-(3-(3,4-dimethylphenyl)-4-oxo-2,3-diazaspiro[4,4]non-l-en-l-
Figure imgf000040_0002
[00122] Prepared in an analogous manner to Example 3 but using 2-(4-(3-(3-(3,4-dimethylphenyl)-4-oxo-2,3- diazaspiro[4,4]non-l-en-l -yl)propyl)phenoxy)-2-methylpropanoic acid (Example 31) in place of 2-(4-(3-( l- (4-(teri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid, and cyclopropylamine in place of benzyl amine as the coupling partners. LC-MS: 502 (M+H)+. Example 33: 2-(4-(3-(3-(3,4-dimethylphenyl -4-oxo-2,3-diazaspiro[4,41non-l-en-l-yl)propyl phenoxy)-2-
Figure imgf000041_0001
[00123] Prepared in an analogous manner to Example 3 but using 2-methyl-2-(4-(3-(4-oxo-3-(4- (trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid (Example 31) in place of 2-(4-(3-(l -(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)- 2-methylpropanoic acid, and 2,2,2-trifluoroethylamine in place of benzyl amine as the coupling partners. LC- MS: 544 (M+H)+.
Example 34: 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl benzyl)-2,3-diazaspiro[4,41non-l-en-l- yl)propyl)phenoxy)propanoic aci
Figure imgf000041_0002
Prepared in an analogous manner to Example 23 but using 4-(trifluoromethyl)benzyl bromide in place of 4- teri-butylbenzyl bromide as the electrophile in step 5. LC-MS: 517 (M+H)+.
Example 35: 2-methyl-2-(4-(3-(4-oxo-3-(3-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,41non-l-en-l- vnpropyl)phenoxy)propanoic acid:
Figure imgf000041_0003
[00124] Prepared in an analogous manner to Example 10 but using 3-iodobenzotrifluoride in place of iodobenzene, and ethyl 2-methyl-2-(4-(3 -(4-oxo-2,3 -diazaspiro [4,4]non- 1 -en- 1 -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l ii-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 503 (M+H)+. Example 36: 2-methyl-2-(4-(3-(4-oxo-3-(3-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,41non-l-en-l- vPpropyl)phenoxy)propanoic acid:
Figure imgf000042_0001
[00125] Prepared in an analogous manner to Example 10 but using 3-(trifluoromethoxy)iodobenzene in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5- dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 519 (M+H)+.
Example 37: 2-methyl-2-(4-(3-(3-(naphthalen-2-yl)-4-oxo-2,3-diazaspiro[4,41non-l-en-l- vnpropyl)phenoxy)propanoic acid:
Figure imgf000042_0002
[00126] Prepared in an analogous manner to Example 10 but using 2-iodonaphthalene in place of iodobenzene, and ethyl 2-methyl-2-(4-(3 -(4-oxo-2,3 -diazaspiro [4,4]non- 1 -en- 1 -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-li7-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 485 (M+H)+.
Example 38: 2-(4-(3-(3-([l,l'-biphenyll-3-yl)-4-oxo-2,3-diazaspiro[4,41non-l-en-l-yl propyl)phenoxy)-2- methylpropanoic acid:
Figure imgf000043_0001
[00127] Prepared in an analogous manner to Example 10 but using 3-iodobiphenyl in place of iodobenzene, and ethyl 2-methyl-2-(4-(3 -(4-oxo-2,3 -diazaspiro[4,4]non- 1 -en- 1 -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoate as coupling partners in step 1. LC-MS: 51 1 (M+H)+.
Example 39: 2-methyl-2-(4-(3-(3-(4-methyl-3-(trifluoromethyl)phenyl)-4-oxo-2,3-diazaspiro[4,41non-l- en-l-yl)propyl)phenoxy)propanoic acid:
Figure imgf000043_0002
[00128] Prepared in an analogous manner to Example 10 but using 5-iodo-2-methylbenzotrifluoride in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5- dihydro-l ii-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 517 (M+H)+.
Example 40: 2-(4-(3-(3-(3-(½rf-butyl)phenyl)-4-oxo-2,3-diazaspiro|4.,4| non-l-en-l-yl)propynphenoxy)-2- methylpropanoic acid:
Figure imgf000043_0003
[00129] Prepared in an analogous manner to Example 10 but using l-(teri-butyl)-3-iodobenzene in place of iodobenzene, and ethyl 2-methyl-2-(4-(3-(4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoate (Example 23, Step 4) in place of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-4,5-dihydro-l //-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoate as coupling partners in step 1. LC-MS: 491 (M+H)+.
Example 41 : 2-methyl-2-(4-(3-(5-oxo-4.|4-dipropyl-l-(4-(trifluoromethyl)phenyl)-4,5-dihvdro-l -
Figure imgf000044_0001
[00130] Step 1: To an ethanol solution (0.2 M) of methyl 2,2-diallyl-6-(4-((l -ethoxy-2 -methyl- 1 -oxopropan- 2-yl)oxy)phenyl)-3-oxohexanoate (1 eq., Example 23, Step 1 ) was added palladium black (0.1 eq., 10% w/w (wet) over carbon). The resulting suspension was evacuated and back-filled with hydrogen gas (3x). Then, the reaction mixture was stirred under a static hydrogen atmosphere maintained with a balloon for 16 h. The mixture was carefully quenched with DCM and filtered through a bed of DCM-wetted celite. The insolubles were then washed further with DCM. Concentration of the filtrate thus obtained in vacuo furnished the desired compound as a colorless oil (93% yield).
[00131] Step 2: To an ethanol solution (0.18 M) of methyl 6-(4-((l -ethoxy-2-methyl-l -oxopropan-2- yl)oxy)phenyl)-3-oxo-2,2-dipropylhexanoate (1 eq.) from the previous step was added hydrazine monohydrate (1.5 eq.). The reaction vessel was then tightly sealed and heated at 120 °C behind a blast shield for 96 h. After cooling to RT, the volatiles were then removed in vacuo and the resulting residue was partitioned between ether and 10% aq. HC1. The aqueous layer was separated and back-extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (5% yield).
[00132] Step 3: To a DMSO solution (0.03 M) of ethyl 2-methyl-2-(4-(3-(5-oxo-4,4-dipropyl-4,5-dihydro- l //-pyrazol-3-yl)propyl)phenoxy)propanoate (1 eq.) from the previous step was added 4-iodobenzotrifluoride (1.5 eq.), copper(I) iodide (0.1 eq.), L-proline (0.2 eq.) and potassium carbonate (3 eq.). The resulting suspension was then deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. Then, the reaction vessel was tightly sealed and the reaction mixture was heated at 100 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil (36% yield).
[00133] Step 4: To a 2: 1 (v/v) THF: MeOH solution (0.1 M) of ethyl 2-methyl-2-(4-(3-(5-oxo-4,4-dipropyl-l - (4-(trifluoromethyl)phenyl)-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)propanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 16 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HCl to pH of ~3. The aqueous suspension thus obtained was then extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04 and filtered. Concentration of the filtrate in vacuo afforded the title compound as a viscous oil (76% yield). LC-MS: 533 (M+H)+.
Example 42: 2-methyl-2-(4-(3-(5-oxo-4,4-dipropyl-l-(3-(trifluoromethyl)phenyl)-4,5-dihydro-lj -
Figure imgf000045_0001
[00134] Prepared in an analogous manner to Example 41 but using 3-iodobenzotrifluoride in place of 4- iodobenzotrifluoride as the aryl iodide coupling partner in step 3. LC-MS: 533 (M+H)+.
Example 43: 4,4-dimethyl-3-(3-(4-((4,4,4-trifluoro-3-hvdroxy-2-methylbutan-2-yl)oxy)phenyl)propyl)-l- (4-(trifluoromethyl)phenvD-lj/-pyrazol-5(4//)-one:
Figure imgf000045_0002
[00135] Step 1: To a THF solution (0.23 M) of 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)phenyl)- 4,5-dihydro-l -pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid (1 eq., Example 19) was added sequentially at 0 °C Hunig's base (1.5 eq.) and ethyl chloroformate (1 .2 eq.). The resulting mixture was then allowed to warm slowly to RT over 16 h. The now white suspension was quenched with methanol (30 eq.) and then added sodium borohydride (6 eq.) portionwise over a period of 5 min. After another 5 h of stirring at RT, the reaction mixture was diluted with 10% aq. HCl and extracted with ether. The combined organic extracts were washed further with 1 N aq. NaOH, water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a viscous oil (90% yield).
[00136] Step 2: To a DCM solution (0.08 M) of 3-(3-(4-(( l -hydroxy-2-methylpropan-2- yl)oxy)phenyl)propyl)-4,4-dimethyl-l-(4-(trifluoromethyl)phenyl)-l /-pyrazol-5(4 /)-one (1 eq.) from the previous step was added sequentially at 0 °C sodium bicarbonate (1.5 eq.) and Dess-Martin periodinane (1.5 eq.). The resulting suspension was then allowed to warm slowly to RT over 3 h before it was diluted with ether and quenched with 5% aq. Na2S203. The organic layer was separated, washed further with 1 N aq. NaOH, water and brine, dried over Na2S04 and filtered. Concentration of the filtrate in vacuo funished the desired product as a viscous oil which was used immediately without further purification.
[00137] Step 3: To a THF solution (0.04 M) of 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromemyl)phenyl)- 4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanal (1 eq.) from the previous step was added sequentially trimethyl(trifluoromethyl)silane (4 eq.) and TBAF (0.2 eq., 1.0 M in THF). The resulting golden yellow solution was then allowed to stir at RT for 16 h before another 1 .5 eq. of TBAF was added. After another 30 min of stirring at RT, the reaction mixture was diluted with ether and washed sequentially with 10% aq. HC1, water and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 4: 1 (v/v) Hex: acetone) afforded the title compound as a pale yellow oil (68% yield over 2 steps). LC-MS: 531 (M+H)+.
Example 44: 4,4-dimethyl-3-(3-(4-((4,4,4-trifluoro-2-methyl-3-oxobutan-2-yl)oxy)phenyl)propylVl-(4-
(trifluoromethyl)phenyl)-l -pyrazol-5(4 f)-one:
Figure imgf000046_0001
[00138] To a DCM solution (0.01 M) of 4,4-dimethyl-3-(3-(4-((4,4,4-trifluoro-3-hydroxy-2-methylbutan-2- yl)oxy)phenyl)propyl)-l-(4-(trifluoromethyl)phenyl)-l//-pyrazol-5(4 /)-one (1 eq., Example 43) was added sequentially at 0 °C sodium bicarbonate (1.5 eq.) and Dess-Martin periodinane (4.5 eq.). The resulting suspension was then allowed to warm slowly to RT over 16 h before it was diluted with ether and quenched with 5% aq. Na2S203. The organic layer was separated, washed further with 1 N aq. NaOH, water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 1 : 1 (v/v) Hex: EtOAc) afforded the title compound as a mixture of the ketone (major) and the corresponding hydrate (minor). LC-MS: 529 (M+H)+. Example 45: 4-(3-(4-((l-amino-2-methylpropan-2-yl oxy phenyl)propyl)-2-(4-(trifluoromethyl phenyl)- 2,3-diazaspiro|4,4|non-3-en-l-o
Figure imgf000047_0001
[00139] Step 1: To a THF solution (0.13 M) of 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3- diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid (1 eq., Example 24) was added sequentially at 0 °C triethylamine (1.5 eq.) and ethyl chloroformate (1.2 eq.). The resulting mixture was then allowed to warm slowly to RT over 16 h. The now pale yellow suspension was quenched with methanol (30 eq.) and then added sodium borohydride (6 eq.) portionwise over a period of 5 min. After another 8 h of stirring at RT, the reaction mixture was diluted with 10% aq. HCl and extracted with ether. The combined organic extracts were washed further with 1 N aq. NaOH, water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 (v/v) Hex: EtOAc -> EtOAc) afforded the desired product as a white foam (78% yield).
[00140] Step 2: To a DCM solution (0.04 M) of 4-(3-(4-(( l -hydroxy-2-methylpropan-2- yl)oxy)phenyl)propyl)-2-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-3-en-l -one (1 eq.) from the previous step was added sequentially at 0 °C triethylamine (1.5 eq.) and methanesulfonyl chloride (1.5 eq.). The resulting suspension was then allowed to warm slowly to RT over 16 h before it was diluted with ether and washed sequentially with cold water, cold 10% aq. HCl and cold brine. The organic layer was then dried over Na2S04 and filtered. Concentration of the filtrate in vacuo funished the desired product as a pale yellow oil which was used immediately without further purification.
[00141] Step 3: To a DMSO solution (0.01 M) of 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3- diazaspiro[4,4]non-l-en-l -yl)propyl)phenoxy)propyl methanesulfonate (1 eq.) from the previous step was added sodium azide (10 eq.). The reaction vessel was then tightly sealed and heated at 140 °C for 16 h behind a blast shield. The resulting dark brown suspension was diluted with ether and washed sequentially with water, 10% aq. HCl, water and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 1 : 1 (v/v) Hex: EtOAc) afforded the title compound as a pale yellow oil (91% yield over 2 steps).
[00142] Step 4: To an ethanol solution (0.01 M) of 4-(3-(4-((l-azido-2-methylpropan-2- yl)oxy)phenyl)propyl)-2-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-3-en-l -one (1 eq.) from the previous step was added palladium black (0.1 eq., 10% w/w (wet) over carbon). The resulting suspension was evacuated and back-filled with hydrogen gas (3x). Then, the reaction mixture was stirred under a static hydrogen atmosphere maintained with a balloon for 3 h. The mixture was carefully quenched with DCM and filtered through a bed of DCM-wetted celite. The insolubles were then washed further with DCM. Concentration of the filtrate thus obtained in vacuo furnished the title compound as a colorless oil (85% yield). LC-MS: 488 (M+H)+.
Example 46: jV-(2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro|4,4| non-1 -en-1-
Figure imgf000048_0001
[00143] To a DCM solution (0.006 M) of 4-(3-(4-((l-amino-2-methylpropan-2-yl)oxy)phenyl)propyl)-2-(4- (trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-3-en-l-one (1 eq., Example 45) was added sequentially triethylamine (1 eq.) and benzenesulfonyl chloride (1 .1 eq.). After 16 h of stirring at RT, the volatiles were then removed in vacuo and the resulting residue was directly subjected to preparative HPLC (gradient elution: 7:3 (v/v) H20: MeCN + 0.1% TFA -> MeCN + 0.1% TFA). The title compound was obtained as a white foam. LC-MS: 628 (M+H)+.
Example 47: V-(2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,41non-l-en-l- vPpropyl)phenoxy)propyl)benzamide:
Figure imgf000048_0002
[00144] Prepared in an analogous manner to Example 46 but using benzoyl chloride in place of benzenesulfonyl chloride as the electrophile. LC-MS: 592 (M+H)+.
Example 48: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihvdro-l -pyrazol-3- yl)propyDphenoxy)-2-phenylacetic acid:
Figure imgf000048_0003
[00145] Step 1: To an acetonitrile solution (0.05 M) of 4-(4-methoxyphenyl)butanoic acid (1 eq.) was added CDI (1.1 eq.). The resulting yellow solution was then allowed to stir at RT for 2.5 h before it was added dropwise, over a period of 1 .5 h, into a white suspension of potassium 3-methoxy-3-oxopropanoate (2.1 eq.), magnesium chloride (2.5 eq.) and triethylamine (3.2 eq.). The resulting suspension was then stirred at RT for 16 h and finally heated at reflux for another 96 h. The crude reaction suspension thus obtained was cooled to RT and diluted with EtOAc. The insolubles were then removed via filtration and rinsed further with EtOAc and DCM. The filtrate thus obtained was concentrated in vacuo, re-taken up in EtOAc and washed sequentially with 10% aq. HC1, water and brine. The organic extract was then dried over Na2S04 and filtered. Concentration of the filtrate in vacuo affored the desired product as an orange oil which can be used without further purification.
[00146] Step 2: To a DMSO solution (0.2 M) of methyl 6-(4-methoxyphenyl)-3-oxohexanoate (1 eq.) from the previous step was added cesium carbonate (3 eq.) and iodomethane (3 eq.). The resulting mixture was then allowed to stir at RT for 16 h. The crude reaction mixture thus obtained was diluted with ether and washed sequentially with cold water, 10% aq. HC1, 1 N aq. NaOH, water and finally brine. The organic extract was then dried over Na2S04, and filtered. Concentration of the filtrate in vacuo affored the desired product as a golden yellow oil which can be used without further purification.
[00147] Step 3: To an ethanol solution (0.75 M) of methyl 6-(4-methoxyphenyl)-2,2-dimethyl-3- oxohexanoate (1 eq.) from the previous step was added hydrazine monohydrate (4 eq.). The reaction vessel was then tightly sealed and heated at 80 °C behind a blast shield for 48 h. After cooling to RT, the volatiles were then removed in vacuo and the resulting residue was partitioned between ether and 10% aq. HC1. The aqueous layer was separated and back-extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04 and filtered. Concentration fo the filtrate in vacuo afforded the desired product as a golden yellow oil which can be used without further purification.
[00148] Step 4: To a DMSO solution (0.13 M) of 3-(3-(4-methoxyphenyl)propyl)-4,4-dimethyl-l i-pyrazol- 5(4ii)-one (1 eq.) from the previous step was added 4-iodobenzotrifluoride (1 .5 eq.), copper(I) iodide (0.1 eq.), L-proline (0.2 eq.) and potassium carbonate (3 eq.). The resulting suspension was then deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. Then, the reaction vessel was tightly sealed and the reaction mixture was heated at 95 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 1 : 1 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil that solidified upon standing (74% yield over 4 steps).
[00149] Step 5: To a DCM solution (0.07 M) of 3-(3-(4-methoxyphenyl)propyl)-4,4-dimethyl-l -(4- (trifluoromethyl)phenyl)-l /-pyrazol-5(4ii)-one (1 eq.) from the previous step was added at -78 °C boron tribromide (3 eq., 1 M in DCM) dropwise over a period of 5 min. The resulting solution was then warmed slowly to RT over 16 h before the reaction was quenched, at -78 °C, with the dropwise addition of methanol. The reaction mixture was then diluted with EtOAc and washed sequentially with 10% aq. HCl, water and brine. The organic extract was dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 4: 1 (v/v) Hex: EtOAc -> EtOAc) afforded the desired product as a pale yellow oil that solidified upon standing (98% yield).
[00150] Step 6: To a DMSO solution (0.07 M) of 3-(3-(4-hydoxyphenyl)propyl)-4,4-dimethyl-l -(4- (trifluoromethyl)phenyl)-l//-pyrazol-5(4//)-one (1 eq.) from the previous step was added cesium carbonate (1.5 eq.) and ethyl 2-bromo-2-phenylacetate (1.2 eq.). The resulting mixture was then stirred at RT for 3 h before the reaction was quenched with the addition of 10% aq. HCl and ether. The organic layer was separated and washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (89% yield).
[00151] Step 7: To a 2: 1 (v/v) THF: MeOH solution (0.024 M) of ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4- (trifluoromethyl)phenyl)-4,5-dihydro-l ii-pyrazol-3-yl)propyl)phenoxy)-2-phenylacetate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 1 .5 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HCl to pH of ~3. The aqueous suspension thus obtained was then extracted with EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, and filtered. Concentration of the filtrate in vacuo, afforded the title compound as a white solid (95% yield). LC-MS: 525 (M+H)+.
Example 49: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l -pyrazol-3- yl)propyDphenoxy)acetic acid:
Figure imgf000050_0001
[00152] Prepared in an analogous manner to Example 48 but using ethyl 2-bromoacetate in place of ethyl 2- bromo-2-phenylacetate as the electrophile in step 6. LC-MS: 449 (M+H)+.
Example 50: l-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l y-pyrazol-3- yl)propyl)phenoxy)cvclobutanecarboxylic acid :
Figure imgf000051_0001
[00153] Prepared in an analogous manner to Example 48 but using ethyl 1-bromocyclobutanecarboxylate in place of ethyl 2-bromo-2-phenylacetate as the electrophile, 100 °C as the reaction temperature and 40 h as the reaction time in step 6. LC-MS: 489 (M+H)+.
Example 51 : l-(4-(3-(4,4-dimethyl-5-oxo-l-(3-(trifluoromethyl)phenyl)-4,5-dihvdro-l -pyrazol-3-
Figure imgf000051_0002
[00154] Prepared in an analogous manner to Example 48 but using 3-iodobenzotrifluoride in place of 4- iodobenzotrifluoride in step 4. Furthermore in step 6, ethyl 1-bromocyclobutanecarboxylate was used in place of ethyl 2-bromo-2-phenylacetate as the electrophile, 120 °C was the optimal reaction temperature and the alkylation was carried out over 24 h. LC-MS: 489 (M+H)+.
Example 52: l-(4-(3-(4.,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihvdro-l -pyrazol-3-
Figure imgf000051_0003
[00155] Prepared in an analogous manner to Example 48 but using ethyl 1 -bromocyclopentanecarboxylate in place of ethyl 2-bromo-2-phenylacetate as the electrophile, 120 °C as the reaction temperature and 56 h as the reaction time in step 6. LC-MS: 503 (M+H)+.
Example 53: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl phenyl)-4,5-dihvdro-l y-pyrazol-3- vPpropyl)phenoxy)-2-phenylpropanoic acid:
Figure imgf000052_0001
[00156] Step 1: To a THF solution (0.05 M) of methyl 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4- (trifluoromethyl)phenyl)-4,5-dihydro-l ii-pyrazol-3-yl)propyl)phenoxy)-2-phenylacetate (1 eq., Example 48, Step 6) was added dropwise KHMDS (1.5 eq., 1 M THF solution) at -78 °C over a period of 5 min. The resulting pink solution was allowed to stir at -78 °C for another 20 min before iodomethane (1.5 eq.) was added neat, dropwise over a period of 2 min. The reaction mixture was then allowed to warm slowly to RT over 16 h. The crude reaction mixture was then diluted with ether washed sequentially withl 0% aq. HC1, water and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -> 3 :7 (v/v) Hex: EtOAc) afforded the racemic product as a pale colorless oil (89% yield). The enantiomers were then be separated on a chiral HPLC (ChiralPak AD column from Daicel Chemical Industries: ID# AD00CJ-D 005, isocratic elution, 1 : 1 (v/v) EtOH: Hexanes, 25 min run). Retention time of Enantiomer 1: 12.2 min. Retention time of Enantiomer 2: 14.7 min.
[00157] Step 2: To a 2: 1 (v/v) THF: MeOH solution (0.019 M) of methyl 2-(4-(3-(4,4-dimethyl-5-oxo-l -(4- (trifluoromethyl)phenyl)-4,5-dihydro-l ii-pyrazol-3-yl)propyl)phenoxy)-2-phenylpropanoate (1 eq.) from the previous step was added lithium hydroxide (3 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 1.5 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 1 N aq. HC1 to pH of ~3. The aqueous suspension thus obtained was then extracted with EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, and filtered. Concentration of the filtrate in vacuo, afforded the title compound as a white solid (Enantiomer 1: 96% yield; Enantiomer 2: 93% yield). LC-MS: 539 (M+H)+.
Example 54: 2-(4-(3-(4,4-dimethyl-5-oxo-l-(3-(trifluoromethyl phenyl)-4,5-dihvdro-l y-pyrazol-3-
Figure imgf000053_0001
[00158] Prepared in an analogous manner to Example 44 but using ethyl 2-(4-(3-(4,4-dimethyl-5-oxo-l -(3- (trifluoromethyl)phenyl)-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2-phenylacetate in place of methyl 2- (4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2- phenylacetate as the substrate in step 1. Furthermore, the chiral separation was carried out using a modified protocol (ChiralPak AD column from Daicel Chemical Industries: ID# AD00CJ-DK.005, isocratic elution, 35 :65 (v/v) EtOH: Hexanes, 15 min run). Retention time of Enantiomer 1: 7.2 min. Retention time of Enantiomer 2: 10.7 min. LC-MS: 539 (M+H)+.
Example 55: 2-(4'-(3-(l-(4-(fg^-butyl benzyl -4,4-dimethyl-5-oxo-4,5-dihvdro-l y-pyrazol-3-yl)propyl)- 4-ethoxy-| l,r-biphenyl|-3-yl)acetic acid:
Figure imgf000053_0002
[00159] Step 1: To an acetonitrile solution (0.14 M) of 4-(4-bromophenyl)butanoic acid (1 eq.) was added CDI (1.1 eq.). The resulting yellow solution was then allowed to stir at RT for 2.5 h before it was added dropwise, over a period of 1.5 h, into a white suspension of potassium 3-methoxy-3-oxopropanoate (2.1 eq.), magnesium chloride (2.5 eq.) and triethylamine (3.2 eq.). The resulting suspension was then stirred at RT for 16 h and finally heated at reflux for another 8 h. The crude reaction suspension thus obtained was cooled to RT and filtered. The filtrate was then concentrated in vacuo and the resulting residue was partitioned between DCM and 20% aq. citric acid. The organic layer was separated and washed further with water and brine, dried over Na2S04 and filtered. Concentration of the filtrate in vacuo affored the desired product as a white solid (33% yield) which can be used without further purification.
[00160] Step 2: To a DMSO solution (0.12 M) of methyl 6-(4-bromophenyl)-3-oxohexanoate (1 eq.) from the previous step was added cesium carbonate (3 eq.) and iodomethane (3 eq.). The resulting mixture was then allowed to stir at RT for 16 h. The crude reaction mixture thus obtained was diluted with ether and washed sequentially with cold water, 10% aq. HC1, 1 N aq. NaOH, water and finally brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -^ 3 :2 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (92% yield).
[00161] Step 3: To an ethanol solution (0.2 M) of methyl 6-(4-bromophenyl)-2,2-dimethyl-3-oxohexanoate (1 eq.) from the previous step was added hydrazine monohydrate (8 eq.). The reaction vessel was then tightly sealed and heated at 50 °C behind a blast shield for 16 h. After cooling to RT, the volatiles were then removed in vacuo and the resulting residue was partitioned between ether and 10% aq. HC1. The aqueous layer was separated and back-extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 3 : 1 (v/v) Hex: acetone) afforded the desired product as a golden yellow oil (92% yield).
[00162] Step 4: To an acetonitrile solution (0.06 M) of 3-(3-(4-bromophenyl)propyl)-4,4-dimethyl-lif- pyrazol-5(4 /)-one (1 eq.) from the previous step was added 4-teri-butylbenzyl bromide (1 .1 eq.) and cesium carbonate (3 eq.). The resulting mixture was then heated at 50 °C for 16 h. After cooling the reaction suspension to RT, the volatiles were removed in vacuo and the resulting residue was partitioned between DCM and 10% aq. HC1. The organic layer was then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -> 3 :2 (v/v) Hex: EtOAc) afforded the desired product as a colorless oil (91 % yield).
[00163] Step 5: To a 3 : 1 (v/v) dioxane: water solution (0.1 M) of 3-(3-(4-bromophenyl)propyl)-l-(4-(teri- butyl)benzyl)-4,4-dimethyl-l /-pyrazol-5(4//)-one (1 eq.) from the previous step was added methyl 2-(2- methoxy-5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)acetate (1.1 eq., prepared according to the patent procedure found in WO/2013/134562), tetrakis(triphenylphosphine)palladium(0) (0.1 eq.) and sodium bicarbonate (15 eq.). The resulting biphasic mixture was deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. The reaction vessel was then tightly sealed and the reaction mixture was heated at 85 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HC1 and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 7:3 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil (60% yield).
[00164] Step 6: To a 2: 1 (v/v) THF: MeOH solution (0.035 M) of methyl 2-(4'-(3-(l -(4-(teri-butyl)benzyl)- 4,4-dimethyl-5-oxo-4,5-dihydro-l ii-pyrazol-3-yl)propyl)-4-ethoxy-[l , l '-biphenyl]-3-yl)acetic acid (1 eq.) from the previous step was added lithium hydroxide (10 eq., 2 N aq. solution). The resulting biphasic mixture was then stirred at RT for 48 h. The volatiles were then removed in vacuo and the resulting residue was acidified with 20% aq. citric acid. The aqueous suspension thus obtained was then extracted with EtOAc. The combined organic extracts were then washed further with water and brine, dried over Na2S04, and filtered. Concentration of the filtrate in vacuo afforded the title compound as a white foam (99% yield). LC-MS: 555 (M+H)+.
Example 56: 2-(4'-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l -pyrazol-3-
Figure imgf000055_0001
[00165] Prepared in an analogous manner to Example 55 but the functionalization of the pyrazalone core (i.e. step 4) is carried out as follows: To a DMSO solution (0.21 M) of 3-(3-(4-bromophenyl)propyl)-4,4-dimethyl- l //-pyrazol-5(4ii)-one (1 eq.) from the previous step was added 4-iodobenzotrifluoride ( 1 .5 eq.), copper(I) iodide (0.1 eq.), L-proline (0.2 eq.) and potassium carbonate (3 eq.). The resulting suspension was then deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. Then, the reaction vessel was tightly sealed and the reaction mixture was heated at 95 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HCI and extracted with ether. The combined organic extracts were then washed further with water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, 9: 1 -^ 3 :7 (v/v) Hex: EtOAc) afforded the desired product as a pinkish solid. LC-MS: 553 (M+H)+.
Example 57: 3-(3-(4'-(3-fluorooxetan-3-yl)-| l , biphenyl|-4-yl)propyl)-4,4-dimethyl-l-(4-
(trifluoromethyl)phenyl)-l -pyrazol-5(4 f)-one:
Figure imgf000055_0002
[00166] To a 2: 1 (v/v) DME: water solution (0.1 1 M) of S-^-^-bromopheny propylM^-dimethyl-l -^- ^rifluoromethy^pheny^-liT-pyrazol-S^/fJ-one (1 eq., Example 56, Step 4) was added 2-(4-(3-fluorooxetan- 3-yl)phenyl)-4,4,5,5-tetramethyl-l ,3,2-dioxaborolane (1.2 eq., prepared according to the patent procedure found in WO/2013/134562), tetrakis(triphenylphosphine)palladium(0) (0.1 eq.) and potassium carbonate (3 eq.). The resulting biphasic mixture was deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. The reaction vessel was then tightly sealed and the reaction mixture was heated at 90 °C for 16 h. After cooling the reaction suspension to RT, the reaction was carefully quenched with 10% aq. HCI and extracted with ether. The combined organic extracts were then washed further with 1 N aq. NaOH, water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 7:3 (v/v) Hex: EtOAc) afforded the title product as a white solid (55% yield). LC-MS: 525 (M+H)+.
Example 58: /V-(6-(4-(3-(l-(4-(fgrf-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihvdro-l -pyrazol-3- yl)propyl)phenvDpyridin-3-yl)benzenesulfonamide:
Figure imgf000056_0001
[00167] Step 1: To a dioxane solution (0.08 M) of 3-(3-(4-bromophenyl)propyl)-l -(4-(fcrt-butyl)benzyl)-4,4- dimethyl-l i-pyrazol-5(4i )-one (1 eq., Example 55, Step 4) was added potassium acetate (3 eq.), bis(pinacolato)diboron (1.3 eq.) and Pd(dppf)Cl2 (0.1 eq.). The resulting pink suspension was deoxygenated via sub-surface purging with a stream of nitrogen for 15 min. The reaction vessel was then tightly sealed and the reaction mixture was heated at 85 °C for 16 h. The volatiles were then removed in vacuo and the resulting residue was partitioned between EtOAc and water. The organic extract was washed further with brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, gradient elution, Hex -^ 3 :2 (v/v) Hex: EtOAc) afforded the desired product as a pale yellow oil (73% yield)
[00168] Step 2: To a 3 : 1 (v/v) dioxane: water solution (0.035 M) of l-(4-(teri-butyl)benzyl)-4,4-dimethyl-3- (3-(4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)propyl)-l i/-pyrazol-5(4ii)-one (1 eq.) from the previous step was added 7V-(6-bromopyridin-3-yl)benzenesulfonamide (1.1 eq.), tetrakis(triphenylphosphine)palladium(0) (0.1 eq.) and sodium bicarbonate (15 eq.). The resulting biphasic mixture was deoxygenated via sub-surface purging with a stream of nitrogen for 5 min. The reaction vessel was then tightly sealed and the reaction mixture was heated at 90 °C for 16 h. After cooling to RT, the reaction mixture was diluted with EtOAc and washed with water and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 45:55 (v/v) Hex: EtOAc) afforded the title product as an off-white solid (45% yield). LC-MS: 609 (M+H)+.
Example 59: N-(6-(4-(3-(l-(4-(fg^-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-lj -pyrazol-3- yl)propyl)phenyl)-3-methoxypyridin-2-yl)benzenesulfonamide:
Figure imgf000057_0001
[00169] Step 1: To a 3 : 1 (v/v) dioxane: water solution (0.036 M) of l -(4-(iert-butyl)benzyl)-4,4-dimethyl-3- (3-(4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)propyl)-l//-pyrazol-5(4//)-one (1 eq., Example 58, step 1) was added 6-bromo-3-methoxy-2-nitropyridine (1.1 eq.), tetrakis(triphenylphosphine)palladium(0) (0.1 eq.) and sodium bicarbonate (15 eq.). The resulting biphasic mixture was deoxygenated via sub-surface purging with a stream of nitrogen for 5 min. The reaction vessel was then tightly sealed and the reaction mixture was heated at 90 °C for 16 h. After cooling to RT, the reaction mixture was diluted with EtOAc and washed with water and brine. The organic extract was then dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 45:55 (v/v) Hex: EtOAc) afforded the desired product as an off-white solid (80% yield).
[00170] Step 2: To a 1 : 1 (v/v) methanol: EtOAc solution (0.02 M) of l -(4-(/ert-butyl)benzyl)-3-(3-(4-(5- methoxy-6-nitropyridin-2-yl)phenyl)propyl)-4,4-dimethyl-l ii-pyrazol-5(4i )-one (1 eq.) from the previous step was added was added palladium black (0.1 eq., 10% w/w (dry) over carbon). The resulting suspension was evacuated and back-filled with hydrogen gas (3x). Then, the reaction mixture was stirred under a static hydrogen atmosphere maintained with a balloon for 16 h. The mixture was carefully quenched with DCM and filtered through a bed of DCM-wetted celite. The insolubles were then washed further with DCM. Concentration of the filtrate thus obtained in vacuo furnished the desired product as an off-white solid (88% yield).
[00171] Step 3: To a pyridine solution (0.04 M) of 3-(3-(4-(6-amino-5-methoxypyridin-2-yl)phenyl)propyl)- l -(4-(/erf-butyl)benzyl)-4,4-dimethyl-lif-pyrazol-5(4ii)-one (1 eq.) from the previous step was added benzenesulfonyl chloride (1.1 eq.). After 16 h of stirring at RT, the volatiles were then removed in vacuo and the resulting residue was taken up in EtOAc. The EtOAc extract was then washed sequentially with sat. aq. CuS04, sat. aq. NaHC03, water and brine, dried over Na2S04, filtered and the filtrate concentrated in vacuo. Purification of the crude product thus obtained by way of column chromatography (Si02, isocratic elution, 3 :2 (v/v) Hex: Acetone) afforded the desired product as a beige solid (16% yield). LC-MS: 639 (M+H)+.

Claims

WHAT IS CLAIMED IS:
1. A compound of Formula I
Figure imgf000058_0001
or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently -Ci_6alkyl, optionally mono-, di- or tri-substituted with halogen, or
R1 and R2 are joined together to form -C3.6cycloalkyl, optionally mono- or di-substituted with substituents independently selected from halogen, -C^alkyl and -CF3;
R3 is selected from the group consisting of:
Figure imgf000058_0002
(b) aryl,
(c) heteroaryl,
Figure imgf000058_0003
wherein the aryl of choice (b) and the heteroaryl of choice (c) are optionally mono- or di-substituted with substituents independently selected from -N(H)R8,
Figure imgf000058_0004
halo-Ci_6alkyl, halo- Ci_6alkoxy, and
Figure imgf000058_0005
-
R4 is selected from the group consisting of:
(a) aryl,
(b) heteroaryl,
(c) -Ci_2alkyl-aryl, and
(d) -C|.2alkyl-heteroaryl,
wherein the aryl of choices (a) and (c), and the heteroaryl of choices (b) and (d), are optionally mono- or di- substituted with substituents selected from the group consisting of halogen, -C^alkyl, -CF3, -OCF3 Ci_ 6alkoxy, -C3.6cycloalkoxy, halo-C|.6alkyl, aryl, heteroaryl, heterocyclo, -C3.6cycloalkyl, and -C3.6cycloalkenyl;
R5 and R6 are each independently selected from the group consisting of: (a) hydrogen,
(b) -C,.6alkyl,
(c) aryl, and
(d) hetereoaryl,
wherein the aryl of choice (c) and heteroaryl of choice (d) are optionally mono- or di-substituted with substituents independently selected from halogen, -Ci_6alkyl and -CF3 or
R5 and R6 are joined together to form a -C3.6cycloalkyl optionally mono- or di-substituted with substituents independently selected from halogen, -d_6alkyl and -CF3;
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3_6cycloalkyl,
(e) -N^S^O C^alkyl,
(f) -N(H)-aryl,
(g) -N(H)-heteroaryl,
(h) -N(H)-C3.6cycloallyl,
(i) -N(H)-C1.6alkyl, and
(j) -CF3>
wherein the aryl of choices (b) and (f), the heteroaryl of choices (c) and (g), the alkyl portion of choices (e) and (i), and the cycloalkyl portion of choices (d) and (h), are optionally mono- or di-substituted with substituents independently selected from halogen, -C|.6alkyl, -CF3, -C3.6cycloalkyl, C|.6alkoxy, C3_ 6cycloalkoxy, aryl, heteroaryl, and hydroxyl; and
R8 is selected from the group consisting of:
(a) hydrogen,
(b) -S(=0)2aryl,
(c) -S(=0)2heteroaryl,
(d) -C(=0)aryl,
(e) -C(=0)heteroaryl,
(f) -S(=0)2-Cwalkyl,
(g) -S(=0)2-C3-6cycloalkyl,
(h) -C(=0)-C,_6alkyl, and
(i) -C(=0)-C3-6cycloalkyl,
wherein the aryl of choices (b) and (d), and the heteroaryl of choices (c) and (e), the alkyl portion of choices (f) and (h), and the cycloalkyl portion of choices (g) and (i), are optionally mono- or di-substituted with substituents independently selected from halogen, -Ci_6alkyl, -CF3, -C3_6cycloalkyl, Ci_6alkoxy, C3_ 6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
2. A compound according to claim 1 or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen or
R1 and R2 are joined together to form -C3.6cycloalkyl, optionally mono or di-substituted with substituents independently selected from halogen, -d_6alkyl and -CF3.
3. A compound according to claim 2 or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen.
4. A compound according to claim 1 of formula la:
Figure imgf000060_0001
la
or a pharmaceutically acceptable salt thereof.
5. A compound according to claim 1 or a pharmaceutically acceptable salt thereof wherein:
R4 is selected from the group consisting of:
(a) -phenyl,
(b) -pyridyl,
(c) -CH2-phenyl, and
(d) -CH2-pyridyl,
wherein the phenyl portion of choices (a) and (c), and the pyridyl portion of choices (b) and (d), are optionally mono- or di-substituted with substituents selected from the group consisting of halogen, -Ci_6alkyl, -CF3, - OCF3, -Ci_6alkoxy, -C3.6cycloalkyl , C3.6cycloalkoxy, and halo-Ci_6alkyl.
6. A compound according to claim 1 or a pharmaceutically acceptable salt thereof wherein:
R5 and R6 are joined together to form a -C3_6cycloalkyl.
7. A compound according to claim 1 or a pharmaceutically acceptable salt thereof wherein:
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3.6cycloalkyl, and
(e) -NCH^^O C^alkyl, wherein the aryl portion of choice (b), the heteroaryl portion of choice (c), the cycloalkyl portion of choice (d) and the alkyl portion of choice (e) are optionally mono- or di -substituted with substituents independently selected from halogen, -Ci_6alkyl, -CF3, -C3.6cycloalkyl, d_6alkoxy, C3.6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
8. A compound according to claim 7 or a pharmaceutically acceptable salt thereof wherein:
R7 is hydroxyl.
9. A compound according to claim 1 or a pharmaceutically acceptable salt thereof wherein:
R8 is selected from the group consisting of:
(a) hydrogen,
(b) -S(=0)2aryl,
(c) -S(=0)2heteroaryl,
(d) -C(=0)aryl, and
(e) -C(=0)heteroaryl,
wherein the aryl of choices (b) and (d), and the heteroaryl of choices (c) and (e), are optionally mono- or di- substituted with substituents independently selected from halogen, -C^alkyl, -CF3, -C3.6cycloalkyl, Ci_ 6alkoxy, C3.6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
A compound according to claim 1 of formula la:
Figure imgf000061_0001
la
or a pharmaceutically acceptable salt thereof wherein:
R1 and R2 are each independently methyl, optionally mono-, di- or tri-substituted with halogen, or
R1 and R2 are joined together to form -C3.6cycloalkyl, optionally mono or di-substituted with substituents independently selected from halogen, -d_6alkyl and -CF3;
R4 is selected from the group consisting of:
(a) -phenyl,
(b) -pyridyl,
(c) -CH2-phenyl, and
(d) -CH2-pyridyl,
wherein the phenyl portion of choices (a) and (c), and the pyridyl portion of choices (b) and (d), are optionally mono- or di-substituted with substituents selected from the group consisting of halogen, -Ci_6alkyl, -CF3, - OCF3, Ci_6alkoxy, -C3_6cycloalkyl , C3.6cycloalkoxy, and halo-Ci_6alkyl; R5 and R6 are joined together to form a -C3_6cycloalkyl; and
R7 is selected from the group consisting of:
(a) hydroxyl,
(b) -N(H)S(=0)2aryl,
(c) -N(H)S(=0)2heteroaryl,
(d) -N(H)S(=0)2-C3.6cycloalkyl, and
Figure imgf000062_0001
wherein the aryl portion of choice (b), the heteroaryl portion of choice (c), the cycloalkyl portion of choice (d) and the alkyl portion of choice (e) are optionally mono- or di -substituted with substituents independently selected from halogen, -Chalky!, -CF3, -C3_6cycloalkyl, -C^alkoxy, C3.6cycloalkoxy, aryl, heteroaryl, and hydroxyl.
1 1. A compound according to claim 10 or a pharmaceutically acceptable salt thereof wherein:
R7 is hydroxyl.
12. A compound according to Claim 1 selected from the group consisting of
2-(4-(3-(l-(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(l-(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2 -methyl-V-(pheny 1 s ulfony 1 )propanami de,
V-benzyl-2-(4-(3-(l -(4-(teri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l ii-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanamide,
2-(4-(3-(l-(4-(ieri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2 -methyl-V-(pyridin-2-ylmethyl)propanamide,
2-(4-(3-(l -(4-(teri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l i-pyrazol-3-yl)propyl)phenoxy)-2-methyl- A^-(pyridin-4-ylmethyl)propanamide,
2-(4-(3-(l -(4-(teri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l i-pyrazol-3-yl)propyl)phenoxy)- V- (cyclohexylmethyl)-2-methylpropanamide,
2-(4-(3 -(4,4-dimethyl-5-oxo- 1 -(3 -(trifluoromemyl)benzyl)-4,5 -dihydro- liT-pyrazol-3 -yl)propyl)phenoxy)-2- methyl propanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)benzyl)-4,5-dihydro-l i-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l-phenethyl-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2 -methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -phenyl-4,5-dihydro-l i-pyrazol-3-yl)propyl)phenoxy)-2-methylpropanoic acid, 2-(4-(3-(l -(4-(teri-butyl)phenyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid, 2-(4-(3-(l-(4-isopropylbenzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-^
methyl propanoic acid,
2-(4-(3-(l -(4-bromobenzyl)-4,4-dirnethyl-5-oxo-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(l-([l,r-biphenyl]-4-ylmemyl)-4,4-dimem^
methylpropanoic acid,
2- (4-(3-(4,4-dimethyl-5-oxo-l-((2\3 \4\5'-tetrahydro^^
3- yl)propyl)phenoxy)-2-methylpropanoic acid,
2-(4-(3-(l -(4-cyclopropylbenzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(l-(4-cyclohexylbenzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2- methyl propanoic acid,
2-(4-(3-(l -(3-fluoro-4-(trifluoromethyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l i-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)phenyl)-4,5-dihydro-li/-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l-(3-(triiluoromethyl)phenyl)-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2- methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -(5-(trifluoromethyl)pyridin-2-yl)-4,5-dihydro-li/-pyrazol-3- yl)propyl)phenoxy)-2-methylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenoxy)-2- methyl- V-(phenylsulfonyl)propanamide,
2-(4-(3-(3-(4-(teri-butyl)benzyl)-4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)-2 -methylpropanoic acid,
2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoic acid,
2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l-en-l-yl)propyl)phenoxy)-N- (pheny 1 s ul fony 1 )propanami de,
V-cyclopropyl-2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanamide,
2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l-en-l-yl)propyl)phenoxy)-N- (2,2,2-trifluoroethyl)propanamide,
2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l-en-l- yl)propyl)phenoxy)propanoic acid,
V-cyclopropyl-2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanamide, 2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l-en-l-yl)propyl)phenV-(2,2,2-trifluoroethyl)propanamide,
2-(4-(3-(3-(3,4-dimethylphenyl)-4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)-2-methylpropanoi acid,
jV-cyclopropyl-2-(4-(3 -(3 -(3 ,4-dimethylphenyl)-4-oxo-2,3 -diazaspiro[4,4]non- 1 -en- 1 -yl)propyl)phenoxy)-2- methylpropanamide,
2-(4-(3-(3-(3,4-dimethylphenyl)-4-oxo-2,3-diazaspiro[4,4]non-l -en-l-yl)propyl)phenoxy)-2-methyl-A^-(2,2,2- trifluoroethyl)propanamide,
2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)benzyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoic acid,
2-methyl-2-(4-(3-(4-oxo-3-(3-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l-en-l- yl)propyl)phenoxy)propanoic acid,
2-methyl-2-(4-(3-(4-oxo-3-(3-(trifluoromethoxy)phenyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoic acid,
2-methyl-2-(4-(3-(3-(naphthalen-2-yl)-4-oxo-2,3-diazaspiro[4,4]non-l -en-l -yl)propyl)phenoxy)propanoic acid,
2-(4-(3 -(3 -([ 1 , Γ -biphenyl]-3 -yl)-4-oxo-2,3 -diazaspiro[4,4]non- 1 -en- 1 -yl)propyl)phenoxy)-2-methylpropanoic acid,
2-methyl-2-(4-(3-(3-(4-methyl-3-(trifluoromethyl)phenyl)-4-oxo-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propanoic acid,
2-(4-(3 -(3 -(3 -( eri-butyl)phenyl)-4-oxo-2,3 -diazaspiro [4,4]non- 1 -en- 1 -yl)propyl)phenoxy)-2-methylpropanoic acid,
2-methyl-2-(4-(3-(5-oxo-4,4-dipropyl-l -(4-(trifluoromethyl)phenyl)-4,5-dihydro-li/-pyrazol-3- yl)propyl)phenoxy)propanoic acid,
2-methyl-2-(4-(3-(5-oxo-4,4-dipropyl-l -(3-(trifluoromethyl)phenyl)-4,5-dihydro-li/-pyrazol-3- yl)propyl)phenoxy)propanoic acid,
4,4-dimethyl-3-(3-(4-((4,4,4-trifluoro-3-hydroxy-2-methylbutan-2-yl)oxy)phenyl)propyl)-l -(4- (tri fl uoromethy 1 )pheny 1 )- 1 //-pyrazol -5 (4H)-o ,
4,4-dimethyl-3-(3-(4-((4,4,4-trifluoro-2-methyl-3-oxobutan-2-yl)oxy)phenyl)propyl)-l -(4- (trifluoromethyl)phenyl)-l /-pyrazol-5(4//)-one,
4-(3 -(4-(( 1 -amino-2-methylpropan-2-yl)oxy)phenyl)propyl)-2-(4-(trifluoromethyl)phenyl)-2,3 - diazaspiro[4,4]non-3-en-l -one,
N-(2-methyl-2-(4-(3 -(4-oxo-3 -(4-(trifluoromethyl)phenyl)-2,3 -diazaspiro[4,4]non- 1 -en- 1 - yl)propyl)phenoxy)propyl)benzenesulfonamide,
V-(2-methyl-2-(4-(3-(4-oxo-3-(4-(trifluoromethyl)phenyl)-2,3-diazaspiro[4,4]non-l -en-l - yl)propyl)phenoxy)propyl)benzamide, 2-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phe
phenylacetic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)phenyl)-4,5-dihydro-li/-pyrazol-3- yl)propyl)phenoxy)acetic acid,
l-(4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)phenyl)-4,5-dihydro-l//-pyrazol-3
yl)propyl)phenoxy)cyclobutanecarboxylic acid,
l-(4-(3-(4,4-dimethyl-5-oxo-l-(3-(trifluoromethyl)phenyl)-4,5-dihydro-l//-pyrazol-3^
yl)propyl)phenoxy)cyclobutanecarboxylic acid,
1 - (4-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)phenyl)-4,5-dihydro-li/-pyrazol-3- yl)propyl)phenoxy)cyclopentanecarboxylic acid,
2- (4-(3-(4,4-dimethyl-5-oxo-l-(4-(trifluoromethyl)ph^
phenylpropanoic acid,
2-(4-(3-(4,4-dimethyl-5-oxo-l -(3-(trifluoromethyl)ph
phenylpropanoic acid,
2-(4'-(3-(l -(4-(ter butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l /-pyrazol-3-yl)propyl)-4-ethoxy-[l , l '- biphenyl]-3-yl)acetic acid,
2- (4'-(3-(4,4-dimethyl-5-oxo-l -(4-(trifluoromethyl)phenyl)-4,5-dihydro-l /-pyrazol-3-yl)propyl)-4-ethox^ [1 , 1 '-biphenyl]-3-yl)acetic acid,
3- (3-(4'-(3-fluorooxetan-3-yl)-[l , l 'biphenyl]-4-yl)propyl)-4,4-dimethyl-l -(4-(trifluoromethyl)phenyl)-lif- pyrazol-5(4H)-one,
A^-(6-(4-(3-(l -(4-(teri-butyl)benzyl)-4,4-dimethyl-5-oxo-4,5-dihydro-l//-pyrazol-3-yl)propyl)phenyl)pyridin- 3-yl)benzenesulfonamide, and
A^-(6-(4-(3-(l -(4-(ie^butyl)benzyl)-4,4-dirnethyl-5-oxo-4,5-dihydro-l i/-pyrazol-3-yl)propyl)phenyl)-3- methoxypyridin-2-yl)benzenesulfonamide,
or a pharaceutically acceptable salt thereof.
13. A pharmaceutical composition comprising a compound according to any one of Claims 1-12, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
14. A method of treating a cancer in a mammal, wherein the cancer is negatively impacted by diminution in its metabolism of fatty acid oxidation, comprising administration of a therapeutically effective amount of a compound of any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, to the mammal in need thereof.
15. The method according to Claim 14, wherein the cancer is selected from prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, and melanoma.
16. A method of treating cancer in a mammal comprising the administration of a therapeutically effective amount of a compound according to any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, to the mammal in need thereof.
17. The method according to Claim 16, wherein the cancer is prostate, breast, ovarian, liver, kidney, colon, pancreatic, human chronic lymphocytic leukemia, or melanoma.
18. A method of treating viral infection in a mammal comprising the administration of a therapeutically effective amount of a compound according to any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, to the mammal in need thereof.
19. The method of treating viral infection of claim 18, wherein the viral infection is hepatitis C virus (HCV) infection or human immunodeficiency virus (HIV) infection.
20. A method of treating a metabolic disorder in a mammal comprising the administration of a therapeutically effective amount of a compound according to any one of Claims 1-12, or a pharmaceutically acceptable salt thereof, to the mammal in need thereof.
21 . A method of preventing the onset of and/or recurrence of cancer in a mammal through the administration of a therapeutically effective amount of a compound according to any one of Claims 1-12, or a pharmaceutically acceptable salt thereof, to the mammal in need thereof.
22. The method of claim 21 , wherein the cancer is a leukemia.
23. The method of claim 22 wherein the cancer is acute or chronic myeloid leukemia.
24. The compound of any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Claim 13, for use in treating cancer in a mammal, wherein said cancer is negatively impacted by diminution in its metabolism of fatty acid oxidation, the use comprising administering a therapeutically effective amount of a compound of any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of Claim 13, to said mammal.
25. The use of a compound of any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, for preparing a medicament for use in treating cancer, comprising the steps of identifying a mammal in need of cancer therapy; wherein said cancer is negatively impacted by diminution in its metabolism of fatty acid oxidation, the use comprising administering a therapeutically effective amount of a compound of any one of Claims 1 -12, or a pharmaceutically acceptable salt thereof, to said mammal.
PCT/US2015/015000 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof WO2015123133A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
BR112016017781A BR112016017781A2 (en) 2014-02-14 2015-02-09 PYRAZOLONE COMPOUNDS AND USES THEREOF
EA201691342A EA201691342A1 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and their use
SG11201606149YA SG11201606149YA (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
US15/118,402 US9862708B2 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
AU2015217450A AU2015217450A1 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
MX2016010436A MX2016010436A (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof.
NZ722345A NZ722345A (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
KR1020167025019A KR20160115996A (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
CA2937542A CA2937542A1 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
CN201580008005.1A CN106029640B (en) 2014-02-14 2015-02-09 Pyrazolinone compounds and application thereof
EP15748977.4A EP3105205A4 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
JP2016552317A JP2017505804A (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
ZA2016/05222A ZA201605222B (en) 2014-02-14 2016-07-27 Pyrazolone compounds and uses thereof
IL247148A IL247148A0 (en) 2014-02-14 2016-08-07 Pyrazolone compounds and uses thereof
US15/829,514 US20180079745A1 (en) 2014-02-14 2017-12-01 Pyrazolone compounds and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461939955P 2014-02-14 2014-02-14
US61/939,955 2014-02-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/118,402 A-371-Of-International US9862708B2 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof
US15/829,514 Continuation US20180079745A1 (en) 2014-02-14 2017-12-01 Pyrazolone compounds and uses thereof

Publications (1)

Publication Number Publication Date
WO2015123133A1 true WO2015123133A1 (en) 2015-08-20

Family

ID=53800550

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/015000 WO2015123133A1 (en) 2014-02-14 2015-02-09 Pyrazolone compounds and uses thereof

Country Status (16)

Country Link
US (2) US9862708B2 (en)
EP (1) EP3105205A4 (en)
JP (1) JP2017505804A (en)
KR (1) KR20160115996A (en)
CN (1) CN106029640B (en)
AU (1) AU2015217450A1 (en)
BR (1) BR112016017781A2 (en)
CA (1) CA2937542A1 (en)
EA (1) EA201691342A1 (en)
IL (1) IL247148A0 (en)
MX (1) MX2016010436A (en)
NZ (1) NZ722345A (en)
PE (1) PE20161315A1 (en)
SG (1) SG11201606149YA (en)
WO (1) WO2015123133A1 (en)
ZA (1) ZA201605222B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021204589A1 (en) 2020-04-07 2021-10-14 Bayer Aktiengesellschaft Substituted thiazolopyridines, salts thereof and their use as herbicidally active substances
WO2022194843A1 (en) 2021-03-19 2022-09-22 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazoles, salts thereof and their use as herbicidally active substances
WO2022229055A1 (en) 2021-04-27 2022-11-03 Bayer Aktiengesellschaft Substituted pyridazinones, salts or n-oxides thereof and their use as herbicidally active substances
WO2023020964A1 (en) 2021-08-17 2023-02-23 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazolyl nicotinamides, salts or n-oxides thereof and their use as herbicidally active substances
WO2023036706A1 (en) 2021-09-07 2023-03-16 Bayer Aktiengesellschaft Substituted thiazolopyridines, salts thereof and their use as herbicidally active substances
WO2023036707A1 (en) 2021-09-07 2023-03-16 Bayer Aktiengesellschaft Substituted 2,3-dihydro[1,3]thiazolo[4,5-b]pyridines, salts thereof and their use as herbicidally active substances
WO2023094594A1 (en) 2021-11-29 2023-06-01 Bayer Aktiengesellschaft Substituted dihydropyranopyridines, salts or n-oxides thereof and their use as herbicidally active substances
EP4238973A1 (en) 2022-03-04 2023-09-06 Bayer AG Substituted 1,2,4-thiadiazolyl isonicotinamides, salts or n-oxides thereof and their use as herbicidally active substances
EP4238972A1 (en) 2022-03-04 2023-09-06 Bayer AG Substituted 1,2,4-thiadiazolyl picolinamides, salts or n-oxides thereof and their use as herbicidally active substances

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3128377A1 (en) * 2019-02-01 2020-08-06 University Of South Carolina Bicyclic pyridine compositions and methods of using the same for cancer therapy

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0121856A2 (en) * 1983-04-08 1984-10-17 Bayer Ag Use of pyrazolone derivatives against the growth of tumour cells and their metastases, medicaments therefor and their preparation
US6455525B1 (en) * 1999-11-04 2002-09-24 Cephalon, Inc. Heterocyclic substituted pyrazolones

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3244593A (en) 1963-06-12 1966-04-05 Endo Lab Pharmaceutically active substituted 5-pyrazolones
US4719175A (en) 1982-05-03 1988-01-12 Polaroid Corporation Novel pyrazole compounds having silver halide development groups
JPH02229168A (en) 1989-03-01 1990-09-11 Takeda Chem Ind Ltd Pyrazolone derivative
EP0491218A1 (en) * 1990-12-17 1992-06-24 F. Hoffmann-La Roche Ag Benzodiazepinones
FR2725988B1 (en) 1994-10-24 1997-01-24 Roussel Uclaf NEW DERIVATIVES OF PYRAZOLONES AND ACID PYRAZOLES, THEIR PROCESS OF PREPARATION, THE NEW INTERMEDIARIES OBTAINED, THEIR APPLICATION AS MEDICINAL PRODUCTS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US5484940A (en) 1994-11-28 1996-01-16 Grant; Francine S. Substituted 3-indolyl-5-pyrazolone compounds
DE19518082A1 (en) 1995-05-17 1996-11-21 Merck Patent Gmbh 4 (-Arylaminomethylene) -2,4-dihydropyrazol-3-one
DE19914140A1 (en) 1999-03-27 2000-09-28 Bayer Ag Substituted benzoylpyrazoles
IL147757A0 (en) 1999-07-30 2002-08-14 Abbott Gmbh & Co Kg 2-pyrazolin-5-ones
US7060822B1 (en) 1999-07-30 2006-06-13 Abbott Gmbh & Co. Kg 2-pyrazolin-5-ones
EP1778669A2 (en) 2004-08-18 2007-05-02 Takeda San Diego, Inc. Kinase inhibitors
AU2005322173A1 (en) 2004-12-27 2006-07-06 Astrazeneca Ab Pyrazolone compounds as metabotropic glutamate receptor agonists for the treatment of neurological and psychiatric disorders
EP1905762A1 (en) 2005-05-30 2008-04-02 Genecare Research Institute Co., Ltd Pyrazolone derivative
WO2006129587A1 (en) 2005-05-30 2006-12-07 Genecare Research Institute Co., Ltd. Pharmaceutical composition comprising pyrazolone derivative
WO2008020150A1 (en) 2006-08-18 2008-02-21 Syngenta Limited Use of hppd inhibitors in the treatment of depression and/or withdrawal symptoms associated with addictive drugs
KR20090031331A (en) 2007-09-21 2009-03-25 주식회사 엘지생명과학 Beta-secretase inhibiting compounds having oxo-dihydro-pyrazole moiety
AU2009318098B2 (en) 2008-11-20 2015-07-16 Northwestern University Treatment of amyotrophic lateral sclerosis
WO2012020738A1 (en) 2010-08-09 2012-02-16 武田薬品工業株式会社 Heterocyclic compound and use thereof
EP2647697B1 (en) 2010-12-01 2016-08-24 Nissan Chemical Industries, Ltd. Method for producing hematopoietic stem cells using pyrazole compound
CA2819215C (en) 2010-12-01 2018-05-01 Nissan Chemical Industries, Ltd. Pyrazole compounds having therapeutic effect on multiple myeloma
WO2013134562A1 (en) * 2012-03-09 2013-09-12 Inception 2, Inc. Triazolone compounds and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0121856A2 (en) * 1983-04-08 1984-10-17 Bayer Ag Use of pyrazolone derivatives against the growth of tumour cells and their metastases, medicaments therefor and their preparation
US6455525B1 (en) * 1999-11-04 2002-09-24 Cephalon, Inc. Heterocyclic substituted pyrazolones

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
See also references of EP3105205A4 *
SHAMSUZZAMAN ET AL.: "Synthesis, evaluation and docking studies on steroidal pyrazolones as anticancer and antimicrobial agents' .", MED. CHEM. RES., vol. 23, January 2014 (2014-01-01), pages 348 - 362, XP055357604 *
WANG, X. -H. ET AL.: "A cell -based screen for anticancer activity of 13 pyrazolone derivatives' .", CHIN. J. CANCER, vol. 29, 2010, pages 980 - 987, XP055372022 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021204589A1 (en) 2020-04-07 2021-10-14 Bayer Aktiengesellschaft Substituted thiazolopyridines, salts thereof and their use as herbicidally active substances
WO2022194843A1 (en) 2021-03-19 2022-09-22 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazoles, salts thereof and their use as herbicidally active substances
WO2022229055A1 (en) 2021-04-27 2022-11-03 Bayer Aktiengesellschaft Substituted pyridazinones, salts or n-oxides thereof and their use as herbicidally active substances
WO2023020964A1 (en) 2021-08-17 2023-02-23 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazolyl nicotinamides, salts or n-oxides thereof and their use as herbicidally active substances
WO2023036706A1 (en) 2021-09-07 2023-03-16 Bayer Aktiengesellschaft Substituted thiazolopyridines, salts thereof and their use as herbicidally active substances
WO2023036707A1 (en) 2021-09-07 2023-03-16 Bayer Aktiengesellschaft Substituted 2,3-dihydro[1,3]thiazolo[4,5-b]pyridines, salts thereof and their use as herbicidally active substances
WO2023094594A1 (en) 2021-11-29 2023-06-01 Bayer Aktiengesellschaft Substituted dihydropyranopyridines, salts or n-oxides thereof and their use as herbicidally active substances
EP4238973A1 (en) 2022-03-04 2023-09-06 Bayer AG Substituted 1,2,4-thiadiazolyl isonicotinamides, salts or n-oxides thereof and their use as herbicidally active substances
EP4238972A1 (en) 2022-03-04 2023-09-06 Bayer AG Substituted 1,2,4-thiadiazolyl picolinamides, salts or n-oxides thereof and their use as herbicidally active substances
WO2023165957A1 (en) 2022-03-04 2023-09-07 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazolyl picolinamides, salts or n-oxides thereof and their use as herbicidally active substances
WO2023165958A1 (en) 2022-03-04 2023-09-07 Bayer Aktiengesellschaft Substituted 1,2,4-thiadiazolyl isonicotinamides, salts or n-oxides thereof and their use as herbicidally active substances

Also Published As

Publication number Publication date
JP2017505804A (en) 2017-02-23
NZ722345A (en) 2018-02-23
MX2016010436A (en) 2016-10-31
US9862708B2 (en) 2018-01-09
EP3105205A4 (en) 2017-08-02
EA201691342A1 (en) 2016-12-30
CN106029640A (en) 2016-10-12
IL247148A0 (en) 2016-09-29
CN106029640B (en) 2018-08-24
SG11201606149YA (en) 2016-09-29
KR20160115996A (en) 2016-10-06
ZA201605222B (en) 2019-02-27
PE20161315A1 (en) 2016-12-25
AU2015217450A1 (en) 2016-08-11
US20170174666A1 (en) 2017-06-22
CA2937542A1 (en) 2015-08-20
US20180079745A1 (en) 2018-03-22
AU2015217450A8 (en) 2016-09-08
EP3105205A1 (en) 2016-12-21
BR112016017781A2 (en) 2017-08-08

Similar Documents

Publication Publication Date Title
US9862708B2 (en) Pyrazolone compounds and uses thereof
US9776976B2 (en) Triazolone compounds and uses thereof
EP2822931B1 (en) Triazolone compounds and uses thereof
US20240041837A1 (en) Triazolone compounds and uses thereof
AU2014232383A1 (en) Heterocyclic compounds useful in the treatment of disease
JPWO2020022237A1 (en) Fused ring lactam derivative
CA3104465A1 (en) Heterocyclic compounds useful in the treatment of disease

Legal Events

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

Ref document number: 15748977

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2937542

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 201691342

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 247148

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 001440-2016

Country of ref document: PE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016017781

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2015217450

Country of ref document: AU

Date of ref document: 20150209

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15118402

Country of ref document: US

Ref document number: MX/A/2016/010436

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2016552317

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: NC2016/0001019

Country of ref document: CO

REEP Request for entry into the european phase

Ref document number: 2015748977

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015748977

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20167025019

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016017781

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160729