WO2023076219A2 - Antagonists of gpr39 protein - Google Patents

Antagonists of gpr39 protein Download PDF

Info

Publication number
WO2023076219A2
WO2023076219A2 PCT/US2022/047658 US2022047658W WO2023076219A2 WO 2023076219 A2 WO2023076219 A2 WO 2023076219A2 US 2022047658 W US2022047658 W US 2022047658W WO 2023076219 A2 WO2023076219 A2 WO 2023076219A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
compound
halogen
pharmaceutically acceptable
Prior art date
Application number
PCT/US2022/047658
Other languages
French (fr)
Other versions
WO2023076219A3 (en
Inventor
Agostino CIANCIULLI
Fabrizio Micheli
Iuni Margaret Laura TRIST
Original Assignee
Vasocardea, 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
Application filed by Vasocardea, Inc. filed Critical Vasocardea, Inc.
Publication of WO2023076219A2 publication Critical patent/WO2023076219A2/en
Publication of WO2023076219A3 publication Critical patent/WO2023076219A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed 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
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D405/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present disclosure concerns novel compounds that act as antagonists to human
  • GPR39 protein is a novel GPR39 protein. Additionally, the present disclosure relates to pharmaceutical compositions and methods of using antagonists to human GPR39 protein in the treatment of diseases or conditions including cardiovascular conditions, endocrine system and hormone disorders, cancer disorders, metabolic diseases, gastrointestinal and liver diseases, hematological disorders, neurological disorders and respiratory diseases.
  • diseases or conditions including cardiovascular conditions, endocrine system and hormone disorders, cancer disorders, metabolic diseases, gastrointestinal and liver diseases, hematological disorders, neurological disorders and respiratory diseases.
  • GPR39 antagonists have been described for methods of treating pain sensitivity, including hyperalgesia, and suppressing appetite (U.S. Pat. Publication 2009/0298756 - Jin et al.).
  • GPR39 agonists and/or antagonists in enhancing glucose regulation and treating impaired carbohydrate metabolism, including in disorders such as diabetes and metabolic syndrome are discussed in WO 2007/141322 - Moreaux et al. - Janssen Pharmaceutica N.V.
  • GPR39 receptor antagonists may be useful in disorders affecting stomach motility, including such as functional dyspepsia and diabetic gastroparesis and/or colorectal motility such as irritable bowel syndrome, diarrhea, or chronic constipation.
  • GPR39 antagonists in the treatment of various cancers is discussed in U.S.
  • Inge Depoortere discloses uses of GPR39 antagonists in treating motility disorders, such as functional dyspepsia, hypoparesis, and chronic constipation in her article, Gl functions of GPR39: novel biology, Current Opinion in Pharmacology, 2012, 12:647-652.
  • the zinc sensing receptor, ZnR/GPR39 controls proliferation and differentiation of colonocytes and thereby tight junction formation in the colon, Cohen et al., Cell Death and
  • GPR antagonists in promoting or enhancing colon epithelial function and tight junction barrier integrity, including treating ulcerative colon diseases, such as ulcerative colitis, and diarrheal pathologies.
  • X 1 is selected from the group of:
  • R a is selected from the group of hydrogen and C 1 -C 3 alkyl;
  • X 2 is selected from the group of: the wavy line in each instance represents a bond through which each X 1 and X 2 moiety is bound;
  • n c is an integer selected from the group of 1,
  • each of R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
  • n1 is an integer selected from the group of 0, 1, 2, and 3;
  • R 2 is selected from the group of carbocycle and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a carbon atom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -(CH 2 ) n1 -C 3 -C 6 cycloalkyl, -CF 3 , halogen, oxo, cyano, -O-(CH 2 ) n2 -C 3 -C 6 cycloalkyl,
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -O-C 1 -C 3 alkyl, with the proviso that R 2 is not pyridinyl unless substituted at least once by -(CH 2 ) n1 -C 3 -C 6 cycloalkyl or phenyl;
  • R 2 ' is selected from the group of hydrogen, OH, halogen, C 1 -C 6 alkyl, and -CF 3 ;
  • R 3 is present one or more times and is independently selected from the group of: a) hydrogen, halogen, cyano, or OH; b) -CO 2 H or -CO 2 -(C 1 -C 6 alkyl); c) C 1 -C 6 alkyl or -O-C 1 -C 6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF 3 , and OH; d) phenyl, benzyl, C 3 -C 6 cycloalkyl, and -CH 2 -C 3 -C 6 cycloalkyl, the rings of each of the phenyl, benzyl, C 3 -C 6 cycloalkyl, and -CH 2 -C 3 -C 6 cycloalkyl groups being substituted by
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF 3 , and -O-C 1 - C 3 alkyl; and, when Y 2 is carbon, R 4 may also be -O-C 1 -C 6 alkyl or two R 4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R 4 C 1
  • OH a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C 1 -C 3 alkyl;
  • R 5 is selected from the group of H and C 1 -C 6 alkyl, wherein the R 5 C 1 -C 6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -
  • R x and R y are independently selected from the group of H and C 1 - C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -
  • R 6 is selected from the group of H, C 1 -C 6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R 6 C 1 -C 6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -NR x R y ,; and OH, and the rings of the R 6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, -CF 3 , and OH, wherein each of R x and R y ; are independently selected from the group of H and C 1 -C 6 alkyl, a heterocycl
  • X 1 is selected from the group of:
  • n a is an integer selected from the group of 0, 1, and 2; n b is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of n a + n b is not less than 2 and not greater than 4; or X 1 and Z 1 together form a fused ring system of the formula (la):
  • R a is selected from the group of hydrogen and C 1 -C 3 alkyl
  • X 2 is selected from the group of: the wavy line in each instance represents a bond through which each X 1 and X 2 moiety is bound;
  • Y 1 is selected from the group of C and N;
  • Y 2 is selected from the group of C, N, S, and O, provided R 4 is not present when Y 2 is O and provided R 4 is either not present or present one or two times when Y 2 is S; with the proviso that no more than one of Y 1 and Y 2 are C;
  • n c is an integer selected from the group of 1, 2, and 3;
  • n d is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of n c + n d is not less than 2 and not greater than 6;
  • Z 1 , Z 2 , and Z 3 are each independently selected from the group of C and N, with the proviso that Z 1 , Z 2 , and Z 3 , when bound to R 2 or non-hydrogen R 2 ' are C;
  • n1 is an integer selected from the group of 0, 1, 2, and 3;
  • R 2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -O-C 1 -C 3 alkyl, with the proviso that R 2 is not pyridinyl unless substituted at least once by -(CH 2 ) n1 -C 3 -C 6 cycloalkyl or phenyl; R 2 ' is selected from the group of OH, halogen, C 1 -C 6 alkyl, and -CF 3 ; R 3 is present one or more times and is independently selected from the group of: g
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF 3 , and -O-C 1 - C 3 alkyl; and, when Y 2 is carbon, R 4 may also be -O-C 1 -C 6 alkyl or two R 4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R 4 C 1
  • OH a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C 1 -C 3 alkyl;
  • R 5 is selected from the group of H and C 1 -C 6 alkyl, wherein the R 5 C 1 -C 6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -
  • R 6 is selected from the group of H, C 1 -C 6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R 6 C 1 -C 6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -NR x R y ,; and OH, and the rings of the R 6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, -CF 3 , and OH, wherein each of R x and R y ; are independently selected from the group of H and C 1
  • X 1 is selected from the group of:
  • n a is an integer selected from the group of 0, 1, and 2; n b is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of n a + n b is not less than 2 and not greater than 4; or X 1 and Z 1 together form a fused ring system of the formula (la):
  • R a is selected from the group of hydrogen and C 1 -C 3 alkyl
  • X 2 is selected from the group of: the wavy line in each instance represents a bond through which each X 1 and X 2 moiety is bound;
  • Y 1 is selected from the group of C and N;
  • Y 2 is selected from the group of C, N, 5, and O, provided R 4 is not present when Y 2 is O and provided R 4 is either not present or present one or two times when Y 2 is 5; with the proviso that no more than one of Y 1 and Y 2 are C;
  • n c is an integer selected from the group of 1, 2, and 3;
  • n d is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of n c + n d 2, 3, 5, or 6;
  • Z 1 , Z 2 , and Z 3 are each independently selected from the group of C and N, with the proviso that Z 1 , Z 2 , and Z 3 , when bound to R 2 or non-hydrogen R 2 ' are C;
  • R 1 is selected
  • each of R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
  • n1 is an integer selected from the group of 0, 1, 2, and 3;
  • R 2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -O-C 1 -C 3 alkyl, with the proviso that R 2 is not pyridinyl unless substituted at least once by -(CH 2 ) n1 -C 3 -C 6 cycloalkyl or phenyl; R 2 ' is selected from the group of hydrogen, OH, halogen, C 1 -C 6 alkyl, and -CF 3 ; R 3 is present one or more times and is independently selected from the group of:
  • OH a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C 1 -C 3 alkyl;
  • R 5 is selected from the group of H and C 1 -C 6 alkyl, wherein the R 5 C 1 -C 6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -
  • R x and R y are independently selected from the group of H and C 1 - C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -
  • R 6 is selected from the group of H, C 1 -C 6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R 6 C 1 -C 6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -NR x R y ,; and OH, and the rings of the R 6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, -CF 3 , and OH, wherein each of R x and R y ; are independently selected from the group of H and C 1
  • a compound of Formula (l-D), or a pharmaceutically acceptable salt thereof wherein: X 1 is selected from the group of: n a is an integer selected from the group of 0, 1, and 2; n b is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of n a + n b is not less than 2 and not greater than 4; or X 1 and Z 1 together form a fused ring system of the formula (la):
  • R a is selected from the group of hydrogen and C 1 -C 3 alkyl
  • X 2 is selected from the group of: the wavy line in each instance represents a bond through which each X 1 and X 2 moiety is bound
  • Y 1 is selected from the group of C and N
  • Y 2 is selected from the group of C, N, S, and O, provided R 4 is not present when Y 2 is O and provided R 4 is either not present or present one or two times when Y 2 is S; with the proviso that no more than one of Y 1 and Y 2 are C
  • n c is an integer selected from the group of 1, 2, and 3
  • n d is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of n c + n d is not less than 2 and not greater than 6
  • Z 1 , Z 2 , and Z 3 are each independently selected from the group of C and N, with the proviso that Z 1 , Z 2 , and Z 3 ,
  • n1 is an integer selected from the group of 0, 1, 2, and 3;
  • R 2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -O-C 1 -C 3 alkyl, with the proviso that R 2 is not pyridinyl unless substituted at least once by -(CH 2 ) n1 -C 3 -C 6 cycloalkyl or phenyl; R 2 ' is selected from the group of hydrogen, OH, halogen, C 1 -C 6 alkyl, and -CF 3 ; R 3 is present one or more times and is independently selected from the group of:
  • R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF 3 , and -O-C 1 - C 3 alkyl; and, when Y 2 is carbon, R 4 may also be -O-C 1 -C 6 alkyl or two R 4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R 4 C 1
  • OH a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C 1 -C 3 alkyl;
  • R 5 is selected from the group of H and C 1 -C 6 alkyl, wherein the R 5 C 1 -C 6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -
  • R x and R y are independently selected from the group of H and C 1 - C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -
  • R 6 is selected from the group of H, C 1 -C 6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R 6 C 1 -C 6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -NR x R y ,; and OH, and the rings of the R 6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, -CF 3 , and OH, wherein each of R x and R y ; are independently selected from the group of H and C 1
  • X 1 is selected from the group of:
  • n a is an integer selected from the group of 0, 1, and 2; n b is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of n a + n b is not less than 2 and not greater than 4; or X 1 and Z 1 together form a fused ring system of the formula (la):
  • R a is selected from the group of hydrogen and C 1 -C 3 alkyl
  • X 2 is selected from the group of: the wavy line in each instance represents a bond through which each X 1 and X 2 moiety is bound;
  • Y 1 is selected from the group of C and N;
  • Y 2 is selected from the group of C, N, 5, and O, provided R 4 is not present when Y 2 is O and provided R 4 is either not present or present one or two times when Y 2 is 5; with the proviso that no more than one of Y 1 and Y 2 are C;
  • n c is an integer selected from the group of 1, 2, and 3;
  • n d is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of n c + n d is not less than 2 and not greater than 6;
  • Z 1 , Z 2 , and Z 3 are each independently selected from the group of C and N, with the proviso that Z 1 , Z 2 , and Z 3 , when bound to R 2 or non-hydrogen R 2 ' are C;
  • each of R x and R y are independently selected from the group of H and C 1 -C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
  • n1 is an integer selected from the group of 0, 1, 2, and 3;
  • R 2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -
  • R 2 is not pyridinyl unless substituted at least once by -(CH 2 ) n1 -C 3 -C 6 cycloalkyl or phenyl;
  • R 2 ' is selected from the group of hydrogen, OH, halogen, C 1 -C 6 alkyl, and -CF 3 ;
  • R 3 is present one or more times and is independently selected from the group of: y) hydrogen, halogen, cyano, or OH; z) -CO 2 H or -CO 2 -(C 1 -C 6 alkyl); aa) C 1 -C 6 alkyl or -O-C 1 -C 6 alkyl substituted by 0,
  • R 4 is present one or two times and is halogen
  • R 5 is selected from the group of H and C 1 -C 6 alkyl, wherein the R 5 C 1 -C 6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -
  • R x and R y are independently selected from the group of H and C 1 - C 6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF 3 , and -
  • R 6 is selected from the group of H, C 1 -C 6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R 6 C 1 -C 6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF 3 , -NR x R y ,; and OH, and the rings of the R 6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, -CF 3 , and OH, wherein each of R x and R y ; are independently selected from the group of H and C 1 -C 6 alkyl, a heterocycl
  • Z 1 , Z 2 , and Z 3 are each C, and R a , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , n a , n b , n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
  • Z 1 , Z 2 , and Z 3 are each N, and R a , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , n a , n b , n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
  • R 7 is present one or two times and is independently selected from the group of hydrogen
  • CF 3 wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
  • R 7 is present one or two times and is independently selected from the group of hydrogen
  • CF 3 wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
  • R 2 is selected from the group of
  • R 7 and R 8 are in each instance independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -CF 3 , and phenyl, with the proviso that only one of R 7 and R 8 may be phenyl.
  • R 7 and R 8 may be bound to any available ring carbon or nitrogen atom in either of the rings and that R 7 and R 8 may be bound to ring atoms in the same or different rings.
  • R 2 is selected from the group of:
  • R 7 and R 8 are in each instance independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, -CF 3 , and phenyl, with the proviso that only one of R 7 and R 8 may be phenyl.
  • separate embodiments comprising, respectfully, compounds for each of the Formulas (l-B), (l-C), (l-D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), or a pharmaceutically acceptable salt thereof, wherein separate embodiments comprise a compound of each formula wherein R 2 is defined above.
  • R 2 is the azepane ring of group a) substituted by variable groups R 7 and R 8 .
  • Another embodiment comprises a compound of Formula (l-B), (l-C), (l-D), or (l-E), wherein R 2 is the azabicyclo[3.10.0]hexanyl ring of group b) substituted by variable groups R 7 and R 8 .
  • R 2 is the azabicyclo[3.10.0]hexanyl ring of group b) substituted by variable groups R 7 and R 8 .
  • each variable including Z 1 , Z 2 , Z 3 , R a , R 1 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , n a , n b , n1, n2, and the provisos are as defined above for the corresponding Formulas (l-B), (l-C), (l-D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R 2 is selected from the group of: wherein R 7 and R 8 are each independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, halogen, and -CF 3 .
  • each variable including Z 1 , Z 2 , Z 3 , R a , R 1 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , n a , n b , n1, n2, and the provisos are as defined above for the corresponding Formula (I), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R 2 is selected from the group of: wherein R 7 and R 8 are each independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, and -CF 3 .
  • each variable including Z 1 , Z 2 , Z 3 , R a , R 1 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , n a , n b , n1, n2, and the provisos are as defined above for the corresponding Formula (I), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R 2 is selected from the group of: wherein R 7 and R 8 are each independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, and -CF 3 .
  • R 2 is selected from the group of: wherein R 7 and R 8 are each independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, and -CF 3 .
  • R 2 is selected from the group of:
  • R 7 and R 8 are each independently selected from hydrogen, halogen, C 1 -C 6 alkyl, -O-C 1 -C 6 alkyl, and -CF 3 .
  • R 7 and R 8 are selected from hydrogen, halogen, -CF 3 , C 1 -C 4 alkyl, and C 1 -C 4 alkoxy. In other embodiments, R 7 and R 8 are selected from hydrogen, halogen, -
  • CF 3 C 1 -C 3 alkyl, and C 1 -C 3 alkoxy.
  • R 3 is selected from the group of hydrogen, C 1 -C 4 alkyl, C 3 - C 6 cycloalkyl, -CF 3 , -C 1 -C 4 alkyl-OH, phenyl, pyrazolyl, and thiophenyl, wherein the phenyl, pyrazolyl, and thiophenyl rings are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, -CF 3 , C 1 -C 4 alkyl, and C 1 -C 4 alkoxy.
  • R 3 is selected from the group of phenyl, pyrazolyl, and thiophenyl, substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, -CF 3 , C 1 -C 4 alkyl, and C 1 -C 4 alkoxy.
  • each compound of Formulas (l-A), (l-B), (l-C), (l-D), and (l-E) and all other formulas and specifically named compounds herein are the pharmaceutically acceptable salts, pharmaceutically acceptable co-crystals, pharmaceutically acceptable esters, pharmaceutically acceptable solvates, hydrates, isomers (including optical isomers, racemates, or other mixtures thereof), tautomers, isotopes, polymorphs, and pharmaceutically acceptable prodrugs of such compounds.
  • the compounds of the disclosure may possess an asymmetric center, and can be produced as a racemic mixture or as individual enantiomers.
  • the individual enantiomers may be obtained by asymmetric synthesis or by resolving a racemic or non-racemic mixture of an intermediate at some appropriate stage of the synthesis.
  • the individual enantiomers may also be obtained by resolution of the compound by conventional means, such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral high pressure liquid chromatography (HPLC) column.
  • HPLC high pressure liquid chromatography
  • BP blood pressure
  • TRH Treatment-resistant hypertension
  • a method of inhibiting the activity of a GPR39 protein in a subject comprising administering to the subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt thereof.
  • the method comprises administering to the subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (I-
  • a method of treating hypertension in a human in need thereof comprising administering to the human a pharmaceutically effective amount of a compound of
  • the hypertension to be treated is primary hypertension, which may also be referred to as essential hypertension.
  • the hypertension to be treated is secondary hypertension, including, but not limited to, hypertension caused by sleep apnea, blocked renal arteries, abnormal levels of hormones, enzymes, growth factors, or other agents controlling systemic or localized blood pressure (including, but not limited to, renin, angiotensin I and II, and aldosterone, angiotensin converting enzyme, catecholamines, thrombin, prostaglandins, natriuretic peptides, vasopressin, adreomedulin, Substance P, calcitonin gene related peptide, kallikreins, kininoogengs, kinins, kinin-degrading enzymes, phosducin, adipokines, and leptin), adrenal gland disease, thyroid abnormalities (including hyperthyroidism, hypothyroidism, Cushing's Disease,
  • the hypertension to be treated is treatment-resistant or refractory hypertension.
  • the hypertension to be treated is hypertensive urgency.
  • the hypertension to be treated is malignant hypertension, which may also be referred to as hypertensive emergency or hypertensive crisis.
  • the hypertension to be treated is isolated systolic hypertension.
  • the hypertension to be treated is salt-sensitive hypertension.
  • the hypertension to be treated is pulmonary hypertension.
  • a method of treating stroke in a human in need thereof comprising administering to the human a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
  • Vascular dementia a small vessel disease in the brain compromises blood flow and oxygen delivery to brain cells, resulting in neuronal dysfunction.
  • Dementia due to small vessel disease is the second leading cause of dementia after Alzheimer's disease, and is the predominant cause in individuals over 70 and in developing countries.
  • a method of treating of dementia due to small vessel disease in a human in need thereof comprising administering to the human a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (I-
  • the compounds and compositions herein may also be used in methods of treating conditions associated with microvascular dysfunction and microvascular disease, with each method comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A),
  • diabetes in question is Type I diabetes mellitus.
  • diabetes in question is Type II diabetes mellitus.
  • prediabetic conditions also known as prediabetes.
  • Also provided is a method of treating heart failure in a human comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (I-
  • cSVD vascular cognitive impairment
  • VCI vascular cognitive impairment
  • Peripheral arterial disease which is mostly small vessel disease and is generally not helped by the surgical and stenting techniques that target large vessels, but do not treat small vessel disease.
  • a method of treatment of peripheral artery disease in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
  • the compounds herein may also be used in the treatment of preeclampsia, the pregnancy complication characterized by high blood pressure and signs of damage to another organ system, most often the liver and kidneys.
  • a method of treatment of preeclampsia in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
  • the compounds herein are also useful in the treatment of myocardial infarction, particularly in cases where no-reflow phenomenon is present.
  • a no-reflow phenomenon is present when, despite restoration of the coronary flow of blood to the myocardium, breakdown of or obstruction to coronary microvasculature can markedly reduce blood flow to the infarct zone. Flow to the microcirculation in such instances may occur in one third of myocardial infarction patients.
  • a method of treatment of myocardial infarction in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
  • the compounds of the present invention may also be used in situations of chronic coronary artery disease in which the large arteries are bypassed or stented and, yet, about a third of the patients continue to have angina due to microvascular dysfunction.
  • a method of treatment of chronic coronary artery disease in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
  • Also provided is a method of treatment of microvascular dysfunction associated with chronic coronary artery disease in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
  • the compounds of the present invention may also be used for conditions referred to as
  • Syndrome X wherein a subject is experiencing a cardiac microvascular dysfunction or constriction causing angina despite normal epicardial coronary artery activity, such as diagnosed by angiography.
  • a method of treatment of Syndrome X in a human comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
  • a method of treating breast cancer in a human in need thereof comprising administering to the human a pharmaceutically effective amount of a compound of
  • the breast cancer treated in this method is ER negative breast cancer.
  • a method of treating gastric adenocarcinomas in a human comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
  • the ulcerative colon disease to be treated is ulcerative colitis.
  • the ulcerative colon disease to be treated is Crohn Disease or Crohn's Disease.
  • the ulcerative colon disease to be treated is Irritable Bowel Syndrome (IBS), which may also be referred to as colitis, enteritis, ileitis, or proctitis.
  • IBS Irritable Bowel Syndrome
  • IBD Inflammatory Bowel Disease
  • a method of treating Inflammatory Bowel Disease (IBD) in a human comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
  • IBD Inflammatory Bowel Disease
  • Also provided is a method of treating diarrhea in a human comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (I-
  • the compounds herein may be used in methods of distributing local, regional, or systemic delivery of anesthetics and improve their effects.
  • a method of enhancing the delivery of anesthetics to a human experiencing microvascular complications comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically effective amount of an anesthetic.
  • the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing, is administered to the human prior to the administration of anesthetic.
  • compositions comprising a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal of any of the foregoing, and a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E) a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal of any of the foregoing, and a pharmaceutically acceptable carrier or excipient.
  • Additional pharmaceutical compositions comprise, respectively, a pharmaceutically effective amount of a compound selected from those of Formulas (I'), (I"), (I'"), (1-1), (1-2), (1-3), (1-4), (1-5), (1-6), and (1-7), and each of the formulas from
  • carrier refers to an excipient or vehicle that includes without limitation diluents, disintegrants, precipitation inhibitors, surfactants, glidants, binders, lubricants, and the like with which the compound is administered.
  • Pharmaceutically acceptable carriers are generally described herein and also in “Remington's Pharmaceutical Sciences” by E. W. Martin.
  • Examples of carriers include, but are not limited to, aluminum monostearate, aluminum stearate, carboxymethylcellulose, carboxymethylcellulose sodium, crospovidone, glyceryl isostearate, glyceryl monostearate, hydroxyethyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxyoctacosanyl hydroxystearate, hydroxypropyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, lactose monohydrate, magnesium stearate, mannitol, microcrystalline cellulose, poloxamer 124, poloxamer 181, poloxamer 182, poloxamer 188, poloxamer 237, poloxamer 407, povidone, silicon dioxide, colloidal silicon dioxide, silicone, silicone adhesive 4102, and silicone emulsion. It should be understood, however, that the carriers selected for the pharmaceutical compositions, and the amounts of such carriers in the composition, may vary depending on the method of
  • excipients in an oral formulation include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • a "pharmaceutically acceptable excipient” is a pharmaceutically acceptable vehicle that includes, without limitation, any and all carriers, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • a pharmaceutically acceptable vehicle that includes, without limitation, any and all carriers, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • compositions may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, for example as described in those patents and patent applications incorporated by reference, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • agents having similar utilities for example as described in those patents and patent applications incorporated by reference, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
  • One mode for administration is parenteral, particularly by injection.
  • Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • Aqueous solutions in saline may also conventionally be used for injection.
  • Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • Sterile injectable solutions are prepared by incorporating a compound according to the present disclosure in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • sterile injectable solutions are prepared containing a therapeutically effective amount, e.g., 0.1 to
  • compositions that include the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co- crystal thereof.
  • Administration may be via capsule or enteric coated tablets, or the like.
  • the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container.
  • a carrier that can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material (as above), which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders.
  • each dosage unit contains from 0.1 mg to 1 g, 0.1 mg to 700 mg, or 0.1 mg to 100 mg of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof.
  • Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt thereof comprises from about 0.1 mg to about 500 mg per dose, given once or twice daily.
  • the individual dose is selected from 1 mg, 5 mg,
  • the amount of the compound actually administered usually will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual subject, the severity of the subject's symptoms, and the like.
  • compositions comprising the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula
  • any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof can be prepared and placed in an appropriate container, and labeled for treatment of an indicated condition. Accordingly, provided is also an article of manufacture, such as a container comprising a unit dosage form of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula
  • the article of manufacture is a container comprising a unit dosage form of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, and at least one pharmaceutically acceptable vehicle.
  • the article of manufacture may be a bottle, vial, ampoule, single-use disposable applicator, or the like, containing the pharmaceutical composition provided in the present disclosure.
  • the container may be formed from a variety of materials, such as glass or plastic and in one aspect also contains a label on, or associated with, the container which indicates directions for use in the treatment of cancer or inflammatory conditions.
  • the active ingredient may be packaged in any material capable of improving chemical and physical stability, such as an aluminum foil bag.
  • diseases or conditions indicated on the label can include, for example, treatment of cancer.
  • Microcirculation may be anatomically defined as blood vessels having a diameter of between about 250 ⁇ tom about 100 , ⁇ pmarticularly including those between about 200 ⁇ m to about 150 ⁇ ,m and includes arterioles, capillaries, and venules (post-capillary venules). Collectively, these vessels may be referred to as "microvessels",
  • microcirculatory vessels and the like.
  • the microcirculation is defined as blood vessels ⁇ 200 ⁇ m in diameter. In the human body approximately 90% of the volume of blood resides in these vessels that include arterioles, capillary and venules. Arterioles range in diameter of approximately from 50-200 ⁇ m.
  • Some venules contain vascular smooth muscle, and some capillaries are surrounded by pericytes, both of which are contractile cells that allow these vessels to constrict and relax, thus allowing more or less blood and oxygen to be delivered to cells, and cell waste removed.
  • pericytes both of which are contractile cells that allow these vessels to constrict and relax, thus allowing more or less blood and oxygen to be delivered to cells, and cell waste removed.
  • the size of these arterioles is tightly regulated to meet the oxygen demand of the cells they supply.
  • Coronary microvascular disease is heart disease that affects the walls and inner lining of tiny blood vessels branching from larger coronary arteries. Coronary microvascular disease may also be referred to as “Cardiac Syndrome X” or “nonobstructive coronary heart disease”. In the heart or elsewhere it may also be referred to as “small artery disease”, “small vessel disease”, or
  • kidney lesions typically involve afferent arterioles and the interlobular artery and may involve intimal thickening, vascular smooth muscle cell proliferation, and extracellular matrix deposition, increasing the media-to-lumen ration. Areas of vascular smooth muscle cells may then be replaced by fibrosis and cell loss.
  • Microvascular angina refers to a form of chest pain due to abnormalities in cardiac microvessels, including, but not limited to, faulty relaxation of or spasms therein.
  • heterocycle or “heterocyclic group” herein refers to a chemical ring containing carbon atoms and at least one ring heteroatom selected from O, S, and N, including saturated, unsaturated, partially saturated, and aromatic rings.
  • 3-membered heterocycles such as seen in the definitions of R 3 and R 4 herein, include by way of example and not limitation aziridinyl, azirinyl, oxiranyl, and thiiranyl groups.
  • Examples of 4-membered heterocycles include by way of example and not limitation azetidinyl, dihydro-1 ⁇ 4 -azetyl, azetyl, 1,3- diazetidinyl, and oxetanyl groups.
  • Examples of 5-membered heterocycles include by way of example and not limitation thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, furanyl, thienyl, pyrrolyl, dihydropyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, pyrrolidinyl, 2-pyrrolidonyl, dihydropyrrolidinyl, pyrrolinyl, tetrahydrofuranyl, triazinyl, thienyl, 2H- pyrrolyl, isothiazolyl, isoxazolyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, and oxazolidinyl groups.
  • 6-membered heterocycles such as seen in the definitions of R 3 and R 4 herein, include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahydropyridyl
  • Examples of 7-membered heterocycles herein include by way of example and not limitation borepinyl, azepanyl, azepinyl, oxepanyl, oxepinyl, theipinyl, thiepanyl, diazepanyl, diazepinyl, and thiazepinyl groups.
  • 8-membered heterocycles herein include by way of example and not limitation azocinyl, azocinyl, oxocanyl, oxocinyl, thiocanyl, and thiocinyl groups.
  • 9-membered heterocycles herein include by way of example and not limitation monocyclic heterocycles such as azonanyl, azoninyl, oxonanyl, oxoninyl, thionanyl, and thioninyl groups and fused heterocycles, such as indolyl, indolinyl, isoindolyl, indolizinyl, indazolyl, azaindolyl, benzimidazolyl, azaindazolyl, pyrazolopyrimidinyl, purinyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, benzo[d]isoxazolyl, benzo[d]isothiazolyl, benzo[d]oxazolyl, benzo[c][l,2,5]thiadiazolyl, benzo[d]thiazolyl, benzisothiazolyl, adeninyl, and
  • 10-membered heterocycles herein include by way of example and not limitation decahydroisoquinolinyl, decahydroquinolinyl, tetrahydroquinolinyl, quinolinyl, dihydroquinolinyl, isoquinolinyl, quinoxalinyl, quinolizinyl, phthalazinyl, quinazolinyl, cinnolinyl, chromenyl, isochromenyl, naphthyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[4,3-d]pyrimidinyl, pyrido[3,4-b]pyrazinyl, pyrido[2,3-b]pyrazinyl, pteridinyl, benzo[e][l,2]oxazinyl, benzo[e][l,3]oxazinyl, benzo[b][l,4]oo
  • heterocycles referenced herein include each isomeric form of the heterocycle, such as the term “dithianyl” including 1,2 dithianyl, 1,3- dithianyl, and 1,4-dithianyl groups, the term “thiadiazinyl” including 1,2,5 thiadiazineyl and 1,3,4- thiadiainyl groups, the term “azaindolyl” including 4-azaindolyl, 5-azaindolyl, 6-azaindolyl, and 7- azaindolyl groups, and "benzothiophenyl” including benzo[b]thiophenyl and benzo[c]thiophenyl groups.
  • heterocycle names include each variance in one or more points of unsaturation.
  • dihydropyrrolyl refers to "2,3-dihydro-lH-pyrrolyl" and
  • alkyl refers to a straight or branched hydrocarbon.
  • an alkyl group can have a specified number of chain carbons, such as 1 to 6 carbon atoms (i.e., C 1 -C 6 alkyl or C 1-6 alkyl).
  • suitable alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl
  • n-butyl --CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l-propyl (i-Bu, i-butyl, --CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s- butyl, --CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, --C(CH 3 ) 3 ), 1-pentyl (n-pentyl, -
  • alkoxy refers to a group having the formula "-O-alkyl,” in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom.
  • the alkyl portion of an alkoxy group can have a specified number of carbon chain atoms, such as 1 to 6 carbon atoms
  • alkoxy groups include, but are not limited to, methoxy (-O-CH 3 or --OMe), ethoxy (-OCH 2 CH 3 or --OEt), t-butoxy (--O--C(CH 3 ) 3 or --OtBu) and the like.
  • Carbocycle or “carbocyclic group” refers to a chemical ring containing only carbon atoms, including saturated, unsaturated, partially saturated, and aromatic rings.
  • cycloalkyl refers to a saturated ring having 3 to 6 carbon atoms as a monocycle, including cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups.
  • halogen refers to an atom selected from the group of elements chlorine, fluorine, bromine, and iodine.
  • therapeutically effective amount and “pharmaceutically effective amount” refer to an amount that is sufficient to effect treatment, as defined below, when administered to a subject (e.g., a mammal, such as a human) in need of such treatment.
  • the therapeutically or pharmaceutically effective amount will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • a “therapeutically effective amount” or a “pharmaceutically effective amount” of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt or cocrystal thereof is an amount sufficient to antagonize GPR39 expression or activity, and thereby treat a subject (e.g., a human) suffering an indication, or to ameliorate or alleviate the existing symptoms of the indication.
  • a therapeutically or pharmaceutically effective amount may be an amount sufficient to decrease a symptom of a disease or condition responsive to inhibition of GPR39 activity.
  • Treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results may include one or more of the following: (i) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); (ii) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread
  • inhibiting indicates a decrease, such as a significant decrease, in the baseline activity of a biological activity or process.
  • Inhibition of GPR39 activity refers to a decrease in GPR39 activity as a direct or indirect response to the presence of a compound of
  • the decrease in activity may be due to the direct interaction of the compound with GPR39, or due to the interaction of the compound(s) described herein with one or more other factors that in turn affect GPR39 expression and/or activity.
  • the presence of the compound(s) may decrease GPR39 activity by directly binding to the GPR39, by causing (directly or indirectly) another factor to decrease GPR39 expression or activity, or by
  • the inhibition of GPR39 activity may be compared in the same subject prior to treatment, or other subjects not receiving the treatment.
  • the term "inhibitor” is understood to refer to a compound or agent that, upon administration to a human in need thereof at a pharmaceutically or therapeutically effective dose, provides the inhibition activity desired.
  • Delaying the development of a disease or condition means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease or condition. This delay can be of varying lengths of time, depending on the history of the disease or condition, and/or subject being treated.
  • a method that "delays" development of a disease or condition is a method that reduces probability of disease or condition development in a given time frame and/or reduces the extent of the disease or condition in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects.
  • Disease or condition development can be detectable using standard methods, such as routine physical exams, mammography, imaging, or biopsy. Development may also refer to disease or condition progression that may be initially undetectable and includes occurrence, recurrence, and onset.
  • significant is meant any detectable change that is statistically significant in a standard parametric test of statistical significance such as Student's T-test, where p ⁇ 0.05.
  • “Pharmaceutically acceptable salts” include, for example, salts with inorganic acids and salts with an organic acid.
  • Examples of salts may include hydrochloride, phosphate, diphosphate, hydrobromide, sulfate, sulfinate, nitrate, malate, maleate, fumarate, tartrate, succinate, citrate, acetate, lactate, methanesulfonate (mesylate), benzenesuflonate (besylate), p-toluenesulfonate
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt particularly a pharmaceutically acceptable addition salt
  • a suitable organic solvent may be used to prepare nontoxic pharmaceutically acceptable addition salts.
  • crystal forms and related terms herein refer to the various crystalline modifications of a given substance, including, but not limited to, polymorphs, solvates, hydrates, co-crystals, and other molecular complexes, as well as salts, solvates of salts, hydrates of salts, other molecular complexes of salts, and polymorphs thereof. Crystal forms of a substance can be obtained by a number of methods, as known in the art.
  • Such methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystallization on surfaces or templates, such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal countermolecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
  • additives such as, e.g., co-crystal countermolecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
  • co-crystal or "co-crystal salt” as used herein means a crystalline material composed of two or more unique solids at room temperature, each of which has distinctive physical characteristics such as structure, melting point, and heats of fusion, hygroscopicity, solubility, and stability.
  • a co-crystal or a co-crystal salt can be produced according to a per se known co-crystallization method.
  • co-crystal or cocrystal salt
  • a host API active pharmaceutical ingredient
  • molecule or molecules such as a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E)
  • guest or co-former
  • the pharmaceutically acceptable co-crystal of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E)with a co-former molecule is in a crystalline form selected from a malonic acid co-crystal, a succinic acid co-crystal, a decanoic acid co-crystal, a salicylic acid co-crystal, a vanillic acid co-crystal, a maltol co-crystal, or a glycolic acid co-crystal.
  • Co-crystals may have improved properties as compared to the parent form (i.e., the free molecule, zwitter ion, etc.) or a salt of the parent compound. Improved properties can include increased solubility, increased dissolution, increased bioavailability, increased dose response, decreased hygroscopicity, a crystalline form of a normally amorphous compound, a crystalline form of a difficult to salt or unsalable compound, decreased form diversity, more desired morphology, and the like.
  • an isotopic compound includes a compound in which one or more hydrogen atoms (H) has been replaced with one or more deuterium atoms (D).
  • deuterium is an isotope of hydrogen, and replacing a hydrogen atom with deuterium (at one or more positions) renders the resulting compound an isotopic compound.
  • isotopic compounds of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), as well as all reference to the various embodiments thereof refers to a compound having one or more isotopic substitutions, including (but not limited to) substitutions of one or more hydrogen atoms with one or more deuterium atoms and any occurrence(s) in the compound.
  • the isotopic compounds disclosed herein provide improved advantages relative to their non- isotopic forms.
  • isotopic modification provides a means of improving existing drugs and/or as a tool in the design of new drugs.
  • isotopic drug design has proven successful in the context of the deuterium (D) kinetic isotope effect.
  • H-D isosteric replacement usually retains the pharmacodynamics of the compound, while improving its pharmacokinetics with a repercussion on half-life and/or of area under the curve values and, ultimately, on dose and/or dosing regimen.
  • drug exposure may be enhanced with isotopic modification, and/or a decrease of clearance.
  • isotopic modification for example, drug exposure may be enhanced with isotopic modification, and/or a decrease of clearance.
  • Subject and patient refer to an animal, such as a mammal, that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in both human therapy and veterinary applications.
  • the subject is a mammal; in some embodiments the subject is human; and in some embodiments the subject is chosen from cats and dogs.
  • Subject in need thereof or “human in need thereof” refers to a subject, such as a human, who may have or is suspected to have diseases or conditions that would benefit from certain treatment; for example treatment with a compound of Formula (l-A),
  • prediabetes or "prediabetic condition” refers to a condition in which a subject's blood sugar levels are not high enough to be considered diabetic but may be a precursor to Type 2 diabetes.
  • a prediabetic condition may be defined in subjects having a fasting blood glucose level of 100 mg/dl or more, but less than 126 mg/dl (the level which is diagnostic for diabetes).
  • Hemoglobin A1c (HbA1c) levels are another laboratory test for diabetes. HbA1c levels of 6.5% or greater are characteristic of diabetes, while levels from 5.7% to 6.4% suggest prediabetes.
  • PC3 Human prostate adenocarcinoma (PC3) cells endogenously expressing human GPR39 receptor were seeded into black walls clear-base 384-well plates at a density of 7,500 cells per well in DM EM (Low Glucose) supplemented with 10% heat-inactivated foetal bovine serum and 1% of
  • Fluorometric Imaging Plate Reader FLIPR TETRA
  • a dual addition FLIPR protocol was used which included a first addition of compounds 200-fold diluted in Assay Buffer containing 150 ⁇ M ZnCI2 and 1.1% DMSO and then after 10 minutes a second addition of a submaximal concentration of the hGPR39 receptor agonist, C3 (Tocris, TC-G
  • f Ki IC50/(l+[L]/EC50) where IC50 is the concentration of antagonist required for 50% inhibition of the maximum response, [L] is the concentration of the agonist used (EC90) and EC50 is the concentration of agonist need to induced 50% of the maximum response (obtained in each experimental plate).
  • fpKi data is shown in Table 1 below. An fpKi less than 5.2 is reported as "+/-”. An fpKi ranging from 5.2 to 5.9 is reported as "+”. An fpKi from 6.0 to 6.5 is reported as "++”. An fpKi from 6.6 to
  • DIPEA N, N- Diisopropylethylamine
  • HATU l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate
  • Compound of formula II may be obtained from compound of formula I (commercially available from
  • Enamine) by SNAr substitution with pyrrolidine in a suitable aprotic solvent e.g. CH 3 CN, typically at
  • Compound of formula III may be obtained by nitro reduction of compound of formula II under catalytic hydrogenation conditions, using for example Pd/C in a suitable solvent such as acetic acid, typically at room temperature. The reaction takes about 4 hours to complete
  • Compound of formula IV may be obtained by coupling reaction between compound of formula III and a proper commercially available acyl chloride, e.g. cyclopropanecarbonyl chloride, in presence of an organic base, e.g. TEA.
  • acyl chloride e.g. cyclopropanecarbonyl chloride
  • organic base e.g. TEA
  • the reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 12 hours to complete.
  • Compound of formula V may be obtained from compound of formula IV by hydrolysis, e.g. using
  • LiOH in a suitable mixture of solvents such as MeOH/THF/H 2 O, typically at 50 °C.
  • the reaction takes about 36 hours to complete.
  • Compound of formula VI may be obtained from compound of formula V by coupling with the proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA.
  • the reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 2-oxa-6-azaspiro[3.4]octane was used instead of 2- phenylpyrrolidine.
  • the title compound was obtained in a yield of 61 % (40 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein trans-2,6-dimethylmorpholine was used instead of 2- phenylpyrrolidine.
  • the title compound was obtained in a yield of 50 % (33 mg) as a racemic mixture.
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 2,6-dimethyl-l,4-thiazinane 1,1-dioxide hydrochloride was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained in a yield of 44 % (34 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 8-oxa-3-azabicyclo[3.2.1]octane hydrochloride was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained in a yield of 44 % (34 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 6-oxa-3-azabicyclo[3.1.1]heptane hydrochloride was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained in a yield of 75 % (49 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-oxa-8-azabicyclo[3.2.1]octane hydrochloride was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained in a yield of 78 % (50 mg).
  • Example 12 The synthesis of the title compound was effected analogously to the synthesis of the compound of
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-[2-(trifluoromethyl)phenyl]piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a yield of 19% (24mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-cyanophenyl)piperazine-l-carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a yield of 47% (55mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(4,5-dimethyl-l,3-thiazol-2-yl)piperazine- 1-carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a yield of 60% (73 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(4-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a yield of 70% (96 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-chloro-5-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a yield of 90% (130 mg).
  • Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine.
  • the title compound was obtained as a racemic mixture in a quantitative yield (150 mg).
  • Compound of formula II may be obtained from compound of formula II by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes about 36 hours to complete.
  • a suitable aprotic solvent e.g. DMSO
  • Compound of formula IV may be obtained from compound of formula III by Buchwald reaction with the proper heteroaryl aniline in the presence of a suitable palladium precatalyst, e.g. tBuBrettPhos Pd G3, and a suitable base, such as K 3 PO 4 .
  • a suitable palladium precatalyst e.g. tBuBrettPhos Pd G3
  • a suitable base such as K 3 PO 4
  • the reaction is carried out in a suitable solvent, e.g. 1- butanol, typically at 120 °C.
  • the reaction takes from about 12 hours to about 24 hours to complete.
  • Compound of formula V may be obtained via Suzuki coupling between compound of formula III and the appropriate commercially available boronic acid in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na 2 CO 3 , in a suitable solvent, 1,2- dimethoxyethane, typically at 95 °C.
  • a suitable catalyst e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na 2 CO 3
  • a suitable solvent 1,2- dimethoxyethane
  • Example 19 The synthesis of the title compound was effected analogously to the synthesis of the compound of
  • Example 18 [4-[(3-methyl-l,2-oxazol-5-yl)amino]-2-pyrrolidin-l-ylphenyl]-(4-methyl-2- phenylpiperazin-l-yl)methanone wherein 5-methyl-l,3,4-thiadiazol-2-amine was used instead of 3- methyl-5-isoxazolamine.
  • the title compound was obtained in a yield of 39 % (56 mg).
  • Example 21 A mixture of (4-bromo-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone (50.0 mg, 0.120 mmol), lH-pyrazole-4-boronic acid (14.37 mg, 0.130 mmol) and a 2M aqueous solution of Na 2 CO 3 (0.18 mL, 0.350 mmol) in 1,2-dimethoxyethane (1.052 mL) was degassed by 5 cycles of nitrogen/vacuum. Then palladium tetrakis triphenylphosphine (6.74 mg, 0.010 mmol) was added and the vessel was sealed. The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and then was heated to 95 °C for 2 days. After this time the mixture was directly purified by FC on
  • Compound of formula II may be obtained from compound of formula I (commercially available from
  • Enamine by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA.
  • the reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
  • compound of formula II may be obtained via acyl chloride formation of compound of formula I (commercially available from Enamine) by treatment with a suitable chlorinating agent, such as SOCl 2 , typically at 90 °C, followed by coupling with the proper amine in the presence of a suitable organic base, e.g. TEA.
  • a suitable chlorinating agent such as SOCl 2
  • TEA a suitable organic base
  • the reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes from about 1 hour to about 12 hours to complete.
  • Compound of formula III may be obtained by nitro reduction of compound of formula II using Zn dust in a suitable solvent, e.g. AcOH, typically at room temperature.
  • a suitable solvent e.g. AcOH
  • the compound of formula III may be obtained by reduction of compound of formula II using Tin (III) chloride dehydrate in a suitable solvent, e.g.
  • EtOH typically at 80 °C.
  • the reaction takes about 12 hours to complete.
  • Compound of formula IV may be obtained by coupling reaction between compound of formula III and a proper commercially available acyl chloride, e.g. Cyclopropanecarbonyl chloride, in presence of an organic base, e.g. TEA.
  • acyl chloride e.g. Cyclopropanecarbonyl chloride
  • organic base e.g. TEA
  • the reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 3 hours to complete.
  • Compound of formula V may be obtained via Suzuki coupling between compound of formula IV and the desired heteroaryl boronic ester or heteroaryl boronic acid, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K 3 PO 4 , in a suitable mixture of solvents, e.g. 1,2- dimethoxyethane / water, typically at 100 °C. The reaction takes about from 3 hours to 12 hours to complete.
  • a suitable precatalyst e.g. XPhos Pd G2
  • a suitable base e.g. K 3 PO 4
  • solvents e.g. 1,2- dimethoxyethane / water
  • Compound of formula VI may be obtained via Miyaura coupling between compound of formula IV, the desired Aryl Bromide, and Bis(pinacolato)diboron, in presence of a suitable catalyst, such as PdCl 2 (dppf)*CH 2 Cl 2 , a suitable base, e.g. AcOK, in a suitable solvent, e.g. 1,4 dioxane, typically at 100
  • Compound of formula V may be obtained via Suzuki coupling between compound of formula VI and commercially available heteroaryl halide in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na 2 CO 3 , in a suitable solvent, 1,2- dimethoxyethane, typically at 95 °C. The reaction takes about 12 hours to complete.
  • a suitable catalyst e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na 2 CO 3
  • a suitable solvent 1,2- dimethoxyethane
  • Example 22 N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein 1-isopropyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole was used instead of l-methyl-lH-pyrazole-3-boronic acid pinacol ester. The title compound was obtained in a yield of 42 % (54 mg).
  • Example 22 N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein N-[3-bromo-4-(4-propylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide and l-isopropyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole were used instead of N-[3-bromo-4-(4,4-difluoropiperidine-l- carbonyl)phenyl]cyclopropanecarboxamide and l-methyl-lH-pyrazole-3-boronic acid pinacol ester respectively.
  • Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein (5-cyanothiophen-2-yl)boronic acid was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2-dioxaborolane. The title compound was obtained in a yield of 11 % (11 mg).
  • Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein [5-(trifluoromethyl)thiophen-2-yl]boronic acid was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2-dioxaborolane. The title compound was obtained in a yield of 11 % (12 mg).
  • Example 30 N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein [5-(trifluoromethyl)thiophen-2-yl]boronic acid was used instead of l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole.
  • the title compound was obtained in a yield of 20 % (11 mg) as a racemic mixture.
  • Example 36 The synthesis of the title compound was effected analogously to the synthesis of the compound of
  • Compound of formula II may be obtained from compound of formula I (commercially available from
  • Astatech by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA.
  • the reaction is carried out in a suitable solvent such as DMF, typically at room temperature.
  • the reaction takes from about 3 hours to about 12 hours to complete.
  • Compound of formula III may be obtained via Suzuki coupling between compound of formula II and the desired heteroaryl boronic ester, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K 3 PO 4 , in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 100 °C.
  • a suitable precatalyst e.g. XPhos Pd G2
  • a suitable base e.g. K 3 PO 4
  • solvents e.g. 1,2-dimethoxyethane / water
  • Compound of formula IV may be obtained by nitrile cyclization of compound of formula III with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
  • Compound of formula V may be obtained by nitrile cyclization of compound of formula III with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
  • Compound of formula IV may be obtained via Suzuki coupling between compound of formula V and the desired heteroaryl boronic ester, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K 3 PO 4 , in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 100 °C.
  • a suitable precatalyst e.g. XPhos Pd G2
  • a suitable base e.g. K 3 PO 4
  • solvents e.g. 1,2-dimethoxyethane / water
  • the title compound was obtained in a yield of 52 % (186 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4-tert-butylpiperidine-l-carbonyl)-3-(l- isopropylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4- methyl-piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 20 % (22 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 3-(l-isopropylpyrazol-3-yl)-4-(l-oxa-7- azaspiro[3.5]nonane-7-carbonyl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 50 % (37 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4-fluoro-4-methylpiperidine-l-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 75 % (32 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-(2-ethylpyrazol-3- yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine- l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 60 % (53 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2- ylpyrazol-4-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 19 % (18 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 39 % (31 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 48 % (50 mg).
  • Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6,6-difluoro-2-azaspiro[3.3]heptane-2-carbonyl)-3-(l- isopropylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4- methyl-piperidine-l-carbonyl)benzonitrile.
  • the title compound was obtained in a yield of 35 % (21 mg).
  • 1H NMR 400 MHz, DMS0-d6) ⁇ 13.73 (s, 1H), 8.30 (d, 1H), 7.93 (d, 1H), 7.84 (d, 1H), 7.33 (d, 1H),

Abstract

Novel compounds that act as antagonists to human GPR39 protein are disclosed. Pharmaceutical compositions and methods of use for antagonists to human GPR39 protein are disclosed. In particular, methods of using the antagonists in the treatment of diseases or conditions including cardiovascular conditions, endocrine system and hormone disorders, cancer disorders, metabolic diseases, gastrointestinal and liver diseases, hematological disorders, neurological disorders and respiratory diseases are disclosed herein.

Description

ANTAGONISTS OF GPR39 PROTEIN
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Patent Application No. 63/272,076, filed on October 26, 2021, the complete disclosure of which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
The present disclosure concerns novel compounds that act as antagonists to human
GPR39 protein. Additionally, the present disclosure relates to pharmaceutical compositions and methods of using antagonists to human GPR39 protein in the treatment of diseases or conditions including cardiovascular conditions, endocrine system and hormone disorders, cancer disorders, metabolic diseases, gastrointestinal and liver diseases, hematological disorders, neurological disorders and respiratory diseases.
BACKGROUND OF THE INVENTION
Use of GPR39 antagonists has been described for methods of treating pain sensitivity, including hyperalgesia, and suppressing appetite (U.S. Pat. Publication 2009/0298756 - Jin et al.).
The use of GPR39 agonists and/or antagonists in enhancing glucose regulation and treating impaired carbohydrate metabolism, including in disorders such as diabetes and metabolic syndrome are discussed in WO 2007/141322 - Moreaux et al. - Janssen Pharmaceutica N.V.
The article Altered Gastrointestinal and Metabolic Function in the GPR39-Obestatin
Receptor-Knockout Mouse, Gastroenterology, Moechars et al., Oct. 2006, Vol. 131, Issue 4, pp.
1131-1141, discloses conclusions that GPR39 receptor antagonists may be useful in disorders affecting stomach motility, including such as functional dyspepsia and diabetic gastroparesis and/or colorectal motility such as irritable bowel syndrome, diarrhea, or chronic constipation.
The role of the obestatin/GPR39 system in human gastric adenocarcinomas, Alen et al.,
Oncotarget 2016, 7:5957-5971 addresses the role of the obestatin/GPR39 system in regulating motility, EMT, and invasion of gastric adenocarcinoma cells. GPR39 antagonists are also discussed in the article Enhanced ZnR/GPR39 Activity in Breast
Cancer, an Alternative Trigger of Signaling Leading to Cell Growth, Ventura-Bixenshpaner et al.,
Scientific Reports (2018) 8:8119, particularly their use in treating breast cancer, including ER negative breast cancer.
The use of GPR39 antagonists in the treatment of various cancers is discussed in U.S.
2004/0071708 (Claassen et al.).
Inge Depoortere discloses uses of GPR39 antagonists in treating motility disorders, such as functional dyspepsia, hypoparesis, and chronic constipation in her article, Gl functions of GPR39: novel biology, Current Opinion in Pharmacology, 2012, 12:647-652.
The article The zinc sensing receptor, ZnR/GPR39, controls proliferation and differentiation of colonocytes and thereby tight junction formation in the colon, Cohen et al., Cell Death and
Disease (2014) 5, e1307, discusses potential uses for GPR antagonists in promoting or enhancing colon epithelial function and tight junction barrier integrity, including treating ulcerative colon diseases, such as ulcerative colitis, and diarrheal pathologies.
BRIEF SUMMARY OF THE INVENTION
Provided is a compound of Formula (l-A), or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0001
wherein: X1 is selected from the group of:
Figure imgf000004_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000004_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl; X2 is selected from the group of:
Figure imgf000005_0001
the wavy line
Figure imgf000005_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, S, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH,
CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3;
R2 is selected from the group of carbocycle and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a carbon atom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl,
-C(=O)OH, -C(=O)-O-C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -
(CH2)n1-heterocyclyl (containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, S, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl;
R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: a) hydrogen, halogen, cyano, or OH; b) -CO2H or -CO2-(C1-C6 alkyl); c) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; d) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; e) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and f) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
Further provided is a compound of Formula (l-B), or a pharmaceutically acceptable salt thereof:
Figure imgf000008_0001
wherein: X1 is selected from the group of:
Figure imgf000008_0002
Figure imgf000009_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000009_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000010_0001
the wavy line
Figure imgf000010_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, S, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: g) hydrogen, halogen, cyano, or OH; h) -CO2H or -CO2-(C1-C6 alkyl); i) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; j) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; k) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and
I) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
Also provided is a compound of Formula (l-C), or a pharmaceutically acceptable salt thereof:
Figure imgf000013_0001
wherein: X1 is selected from the group of:
Figure imgf000013_0002
Figure imgf000014_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000014_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000014_0003
the wavy line
Figure imgf000015_0001
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, 5, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is 5; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd 2, 3, 5, or 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH,
CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: m) hydrogen, halogen, cyano, or OH; n) -CO2H or -CO2-(C1-C6 alkyl); o) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; p) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; q) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and r) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-
C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
Further also provided is a compound of Formula (l-D), or a pharmaceutically acceptable salt thereof:
Figure imgf000017_0001
wherein: X1 is selected from the group of:
Figure imgf000018_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000018_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl; X2 is selected from the group of:
Figure imgf000019_0001
the wavy line
Figure imgf000019_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, S, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: s) hydrogen, halogen, cyano, or OH; t) -CO2H or -CO2-(C1-C6 alkyl); u) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; v) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; w) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and x) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
Further yet provided is a compound of Formula (l-E), or a pharmaceutically acceptable salt thereof:
Figure imgf000022_0001
wherein: X1 is selected from the group of:
Figure imgf000022_0002
Figure imgf000023_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000023_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000023_0003
the wavy line
Figure imgf000024_0001
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, 5, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is 5; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH,
CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: y) hydrogen, halogen, cyano, or OH; z) -CO2H or -CO2-(C1-C6 alkyl); aa) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; bb) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; cc) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and dd) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is halogen;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3, with the proviso that the compound is not N-[4-(4,4-difluoropiperidine-l-carbonyl)-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide; (4,4-difluoropiperidin-l-yl)-[4-(5-methyl-4H- l,2,4-triazol-3-yl)-2-[3-(trifluoromethyl)pyrazol-l-yl]phenyl]methanone; [2-(3-cyclopropylpyrazol- l-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone; (4,4- difluoropiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(3-propan-2-ylpyrazol-l- yl)phenyl]methanone; or N-[4-(4-fluoropiperidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide.
DETAILED DESCRIPTION OF THE INVENTION
Within the scope of compounds of Formula (l-A), (l-B), (l-C), (l-D) and (l-E), or pharmaceutically acceptable salts thereof, there are three additional embodiments comprising, respectively, a compound of Formula (I') or a pharmaceutically acceptable salt thereof, a compound of Formula (I") or a pharmaceutically acceptable salt thereof, and a compound of
Formula (I'") or a pharmaceutically acceptable salt thereof:
Figure imgf000026_0001
wherein, in each instance, when present, X1, X2, R2, Z1, Z2, Z3, and all other variables and provisos are as defined for Formulas (l-A), (l-B), (l-C), (l-D), or (l-E), above.
It is also understood that there are separate and independent embodiments within the scope of Formula (I) and based upon the definitions of X1 and X2, above, comprising, respectively, a compound of Formulas (1-1), (1-2), (1-3), (1-4), (1-5), (1-6), and (1-7), or a pharmaceutically acceptable salt thereof:
Figure imgf000027_0001
Figure imgf000028_0001
wherein, in each instance in which they are present, all variables, including X1, Ra, R1, R2, R3, R4, Y1, Y2, Z1, Z2, Z3, na, nb, n1, n2, and the provisos are as defined above for Formulas (l-A), (l-B), (l-C), (I-
D), or (l-E), .
It is also understood that there are separate and independent embodiments within the scope of Formulas (l-A), (l-B), (l-C), (l-D), and (l-E), and based upon the definitions of X1 and X2, above, comprising, respectively, a compound of Formulas (l-a) through (l-ll), below, or a pharmaceutically acceptable salt thereof:
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
wherein, in each instance in which they are present, all variables, including Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, Z1, Z2, Z3, na, nb, n1, n2, and the provisos are as defined above for Formulas (l-A), (l-B), (l-C),
(l-D), or (l-E).
Other separate embodiments provide a compound of each of Formulas (l-A), (l-B), (l-C), (I-
D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (l-l) through (I-7), and Formulas (l-a) through (I-
II), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment Z1, Z2, and Z3 are each C, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Other separate embodiments provide a compound of each of Formulas (l-A), (l-B), (l-C), (I-
D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (I-
II), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment Z1, Z2, and Z3 are each N, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Additional separate embodiments provide a compound of each of Formulas (l-A), (l-B), (I-
C), (l-D), and (l-E), Formulas (I'), (I"), and (I"'), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment, Z1 and Z2 are each C, Z3 is N, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-;ll). Additional separate embodiments provide a compound of each of Formulas (l-A), (l-B), (I-
C), (l-D), and (l-E), Formulas (I'), (I"), and (I"'), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment, Z1 and Z3 are each C, Z2 is N, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Additional separate embodiments provide a compound of each of Formulas (l-A), (l-B), (I-
C), (l-D), and (l-E), Formulas (I'), (I"), and (I"'), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment, Z1 and Z3 are each N, Z2 is C, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Additional separate embodiments provide a compound of each of Formulas (l-A), (l-B), (I-
C), (l-D), and (l-E), Formulas (I'), (I"), and (I"'), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment, Z1 and Z2 are each N, Z3 is C, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Additional separate embodiments provide a compound of each of Formulas (l-A), (l-B), (I-
C), (l-D), and (l-E), Formulas (I'), (I"), and (I"'), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, wherein in each embodiment, Z2 and Z3 are each N, Z1 is C, and Ra, R1, R2, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-A), (l-B), (l-C), (l-D), or (l-E) through (l-ll).
Within each of the embodiments herein of Formula (l-A), Formulas (I'), (I"), and (I'"),
Formulas (1-1) through (1-7), and Formulas (l-a) through (l-jj), above, or a pharmaceutically acceptable salt thereof, there is a further embodiment in which R2 is selected from the group of
Figure imgf000036_0001
wherein R7 is present one or two times and is independently selected from the group of hydrogen,
-OH; halogen; -C1-C6 alkyl; -O-C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -
C(=O)OH; -C(=O)-O-C1-C6 alkyl; -S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -
CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
Within each of the embodiments herein of Formula (l-A), Formulas (I'), (I"), and (I'"),
Formulas (1-1) through (1-7), and Formulas (l-a) through (l-jj), above, or a pharmaceutically acceptable salt thereof, there is a further embodiment in which R2 is selected from the group of
Figure imgf000037_0001
Figure imgf000038_0001
wherein R7 is present one or two times and is independently selected from the group of hydrogen,
-OH; halogen; -C1-C6 alkyl; -O-C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -
C(=O)OH; -C(=O)-O-C1-C6 alkyl; -S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -
CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
Within each of the embodiments herein of Formula (l-A), Formulas (I'), (I"), and (I'"),
Formulas (1-1) through (1-7), and Formulas (l-a) through (l-jj), above, or a pharmaceutically acceptable salt thereof, there is yet a further embodiment in which R2 is selected from the group of
Figure imgf000039_0002
Within each of the embodiments herein of Formulas (l-B), (l-C), (l-D), and (l-E), Formulas
(I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, there is a further embodiment in which R2 is selected from the group of
Figure imgf000039_0001
Figure imgf000040_0001
wherein R7 and R8 are in each instance independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, -CF3, and phenyl, with the proviso that only one of R7 and R8 may be phenyl.
It is understood that in the embodiments herein, in instances wherein the R2 group is a bicyclic or spirocyclic ring, R7 and R8 may be bound to any available ring carbon or nitrogen atom in either of the rings and that R7 and R8may be bound to ring atoms in the same or different rings.
Within each of the embodiments herein of Formulas (l-B), (l-C), (l-D), and (l-E), Formulas
(I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), above, or a pharmaceutically acceptable salt thereof, there is yet a further embodiment in which R2 is selected from the group of:
Figure imgf000040_0002
Figure imgf000041_0001
wherein R7 and R8 are in each instance independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, -CF3, and phenyl, with the proviso that only one of R7 and R8 may be phenyl.
Also provided are separate embodiments comprising, respectfully, compounds for each of the Formulas (l-B), (l-C), (l-D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), or a pharmaceutically acceptable salt thereof, wherein separate embodiments comprise a compound of each formula wherein R2 is defined above. For instance, provided is a compound of Formula (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt thereof, wherein R2 is the azepane ring of group a) substituted by variable groups R7 and R8.
Another embodiment comprises a compound of Formula (l-B), (l-C), (l-D), or (l-E), wherein R2 is the azabicyclo[3.10.0]hexanyl ring of group b) substituted by variable groups R7 and R8. This pattern continues to define the remainder of the embodiments, each with the separate referenced R2 group from the list above.
Within each of the embodiments above for a compound of each of Formulas (l-B), (l-C), (I-
D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through
(I-II), or a pharmaceutically acceptable salt thereof, there is also a further embodiment wherein each variable, including Z1, Z2, Z3, Ra, R1, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formulas (l-B), (l-C), (l-D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R2 is selected from the group of:
Figure imgf000042_0001
wherein R7 and R8 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, halogen, and -CF3.
Within each of the embodiments above for a compound of each of Formulas (l-B), (l-C), (I-
D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through
( l-ll), or a pharmaceutically acceptable salt thereof, there is also a further embodiment wherein each variable, including Z1, Z2, Z3, Ra, R1, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formula (I), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R2 is selected from the group of:
Figure imgf000042_0002
wherein R7 and R8 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
Within each of the embodiments above for a compound of each of Formulas (l-B), (l-C), (I-
D), and (l-E), Formulas (I'), (I"), and (I'"), Formulas (1-1) through (1-7), and Formulas (l-a) through
(l-ll), or a pharmaceutically acceptable salt thereof, there is also a further embodiment wherein each variable, including Z1, Z2, Z3, Ra, R1, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formula (I), Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R2 is selected from the group of:
Figure imgf000043_0001
wherein R7 and R8 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
Within each of the embodiments above for a compound of each of Formula (I), Formulas
(1-1) through (1-7), and Formulas (l-a) through (l-ll), or a pharmaceutically acceptable salt thereof, there is also a further embodiment wherein each variable, including Z1, Z2, Z3, Ra, R1, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formula (I),
Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R2 is selected from the group of:
Figure imgf000043_0002
wherein R7 and R8 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
Within each of the embodiments above for a compound of each of Formula (I), Formulas
(1-1) through (1-7), and Formulas (l-a) through (l-ll), or a pharmaceutically acceptable salt thereof, there is also a further embodiment wherein each variable, including Z1, Z2, Z3, Ra, R1, R3, R4, R5, R6, Y1, Y2, na, nb, n1, n2, and the provisos are as defined above for the corresponding Formula (I),
Formulas (1-1) through (1-7), and Formulas (l-a) through (l-ll), and R2 is selected from the group of:
Figure imgf000044_0001
wherein R7 and R8 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
In some embodiments herein, R7 and R8 are selected from hydrogen, halogen, -CF3, C1-C4 alkyl, and C1-C4 alkoxy. In other embodiments, R7 and R8 are selected from hydrogen, halogen, -
CF3, C1-C3 alkyl, and C1-C3 alkoxy. In still further embodiments, hydrogen, F, Cl, , -CF3, C1-C3 alkyl, and C1-C3 alkoxy.
Within each of the embodiments for compounds and salts herein comprising a R3 moiety, there is a further embodiment of the same scope, except R3 is present one to three times. Within each of the embodiments for compounds and salts herein comprising a R3 moiety, there is a further embodiment of the same scope, except R3 is present one to two times. Within each of the embodiments for compounds and salts herein comprising a R3 moiety, there is a further embodiment of the same scope, except R3 is present only one time. Within each of the embodiments for compounds and salts herein comprising a R3 moiety, there is a further embodiment of the same scope, except R3 is selected from the group of hydrogen, C1-C4 alkyl, C3- C6 cycloalkyl, -CF3, -C1-C4 alkyl-OH, phenyl, pyrazolyl, and thiophenyl, wherein the phenyl, pyrazolyl, and thiophenyl rings are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, -CF3, C1-C4 alkyl, and C1-C4 alkoxy.
Within each of the embodiments for compounds and salts herein comprising a R3 moiety, there is a further embodiment of the same scope, except R3 is selected from the group of phenyl, pyrazolyl, and thiophenyl, substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, -CF3, C1-C4 alkyl, and C1-C4 alkoxy.
Also included for each compound of Formulas (l-A), (l-B), (l-C), (l-D), and (l-E) and all other formulas and specifically named compounds herein are the pharmaceutically acceptable salts, pharmaceutically acceptable co-crystals, pharmaceutically acceptable esters, pharmaceutically acceptable solvates, hydrates, isomers (including optical isomers, racemates, or other mixtures thereof), tautomers, isotopes, polymorphs, and pharmaceutically acceptable prodrugs of such compounds.
The compounds of the disclosure may possess an asymmetric center, and can be produced as a racemic mixture or as individual enantiomers. The individual enantiomers may be obtained by asymmetric synthesis or by resolving a racemic or non-racemic mixture of an intermediate at some appropriate stage of the synthesis. The individual enantiomers may also be obtained by resolution of the compound by conventional means, such as crystallization in the presence of a resolving agent, or chromatography, using, for example a chiral high pressure liquid chromatography (HPLC) column. The individual enantiomers as well as racemic and non-racemic mixtures of enantiomers are within the scope of the present disclosure, all of which are intended to be included within the structures depicted in this specification unless otherwise specifically indicated.
Methods of Use
Hypertension is the most common chronic disease, affecting 1.13 billion people and accounting for 10 million deaths every year worldwide. Despite the availability of antihypertensive drugs that successfully reduced blood pressure (BP) for millions of patients, important unmet needs remain. Only half of people with hypertension have their BP under control, and current antihypertensive medications fail to restore normal BP in a large proportion of patients.
Treatment-resistant hypertension (TRH), also known as resistant hypertension, is on the rise due to the rising rates of diabetes and obesity, which contribute up to 75% of the risk for hypertension and to most cases of TRH. Identifying new targets and developing novel strategies for preventing and treating hypertension and its complications remain a high public health and medical priority.
Provided is a method of inhibiting the activity of a GPR39 protein in a subject, the method comprising administering to the subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt thereof.
That said, in certain embodiments, the method comprises administering to the subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (I-
D), or (l-E), of a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), of any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Provided is a method of treating hypertension in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound of
Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C),
(l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing. In one embodiment, the hypertension to be treated is primary hypertension, which may also be referred to as essential hypertension.
In another embodiment, the hypertension to be treated is secondary hypertension, including, but not limited to, hypertension caused by sleep apnea, blocked renal arteries, abnormal levels of hormones, enzymes, growth factors, or other agents controlling systemic or localized blood pressure (including, but not limited to, renin, angiotensin I and II, and aldosterone, angiotensin converting enzyme, catecholamines, thrombin, prostaglandins, natriuretic peptides, vasopressin, adreomedulin, Substance P, calcitonin gene related peptide, kallikreins, kininoogengs, kinins, kinin-degrading enzymes, phosducin, adipokines, and leptin), adrenal gland disease, thyroid abnormalities (including hyperthyroidism, hypothyroidism, Cushing's Disease,
Pheochormocytoma, and excess growth hormone), and constrictions of the aorta, including coarctation.
In one embodiment, the hypertension to be treated is treatment-resistant or refractory hypertension.
In still another embodiment, the hypertension to be treated is hypertensive urgency.
In another embodiment, the hypertension to be treated is malignant hypertension, which may also be referred to as hypertensive emergency or hypertensive crisis.
In an additional embodiment, the hypertension to be treated is isolated systolic hypertension.
In a further embodiment, the hypertension to be treated is salt-sensitive hypertension.
In another embodiment, the hypertension to be treated is pulmonary hypertension.
Current therapies for stroke involve dissolving or removing the clot responsible for vessel occlusion. Yet, despite opening large vessels, small blood vessels, remain compromised in a significant proportion of patients, ranging from 30-50%, which can lead to ineffective therapy. A compound as described herein can be combined with current stroke therapy to improve outcome for a large segment of patients.
Accordingly, provided is a method of treating stroke in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Vascular dementia a small vessel disease in the brain compromises blood flow and oxygen delivery to brain cells, resulting in neuronal dysfunction. Dementia due to small vessel disease is the second leading cause of dementia after Alzheimer's disease, and is the predominant cause in individuals over 70 and in developing countries. Provided is a method of treating of dementia due to small vessel disease in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (I-
E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
In light of their effects on relaxing and protecting microvessels, the compounds and compositions herein may also be used in methods of treating conditions associated with microvascular dysfunction and microvascular disease, with each method comprising administering to a subject in need thereof a pharmaceutically effective amount of a compound of Formula (l-A),
(l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing. This includes methods of treating microvascular complications associated with or caused by diabetes. In one embodiment, the diabetes in question is Type I diabetes mellitus. In another embodiment the diabetes in question is Type II diabetes mellitus. This also includes methods of treating microvascular complications associated with or caused by prediabetic conditions, also known as prediabetes.
Also provided is a method of treating heart failure in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (I-
C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing. Included are methods of treating microvascular angina, cerebral small vessel disease
(cSVD), vascular cognitive impairment (VCI), Systemic Microvascular Endothelial Dysfunction
(alone or presenting with Infective Endocarditis), and Chronic Kidney Disease.
Peripheral arterial disease, which is mostly small vessel disease and is generally not helped by the surgical and stenting techniques that target large vessels, but do not treat small vessel disease.
Provided herein is a method of treatment of peripheral artery disease in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
The compounds herein may also be used in the treatment of preeclampsia, the pregnancy complication characterized by high blood pressure and signs of damage to another organ system, most often the liver and kidneys. Provided herein is a method of treatment of preeclampsia in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
The compounds herein are also useful in the treatment of myocardial infarction, particularly in cases where no-reflow phenomenon is present. A no-reflow phenomenon is present when, despite restoration of the coronary flow of blood to the myocardium, breakdown of or obstruction to coronary microvasculature can markedly reduce blood flow to the infarct zone. Flow to the microcirculation in such instances may occur in one third of myocardial infarction patients.
As such, provided is a method of treatment of myocardial infarction in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Also provided is a method of treatment of myocardial infarction with the presence of noreflow phenomenon in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or
(l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
The compounds of the present invention may also be used in situations of chronic coronary artery disease in which the large arteries are bypassed or stented and, yet, about a third of the patients continue to have angina due to microvascular dysfunction.
As such, provided is a method of treatment of chronic coronary artery disease in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Also provided is a method of treatment of microvascular dysfunction associated with chronic coronary artery disease in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
The compounds of the present invention may also be used for conditions referred to as
Syndrome X, wherein a subject is experiencing a cardiac microvascular dysfunction or constriction causing angina despite normal epicardial coronary artery activity, such as diagnosed by angiography. As such, provided is a method of treatment of Syndrome X in a human, the method comprising administering to a mammal in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Provided is a method of treating breast cancer in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound of
Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C),
(l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing. In some embodiments, the breast cancer treated in this method is ER negative breast cancer. Also provided is a method of treating gastric adenocarcinomas in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (I-
A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Also provided is a method of promoting or enhancing colon epithelial function and tight junction barrier integrity in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or
(l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Similarly provided is a method of treating ulcerative colon diseases in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing. In one embodiment, the ulcerative colon disease to be treated is ulcerative colitis. In another embodiment, the ulcerative colon disease to be treated is Crohn Disease or Crohn's Disease. In still another embodiment, the ulcerative colon disease to be treated is Irritable Bowel Syndrome (IBS), which may also be referred to as colitis, enteritis, ileitis, or proctitis.
Also provided is a method of treating Inflammatory Bowel Disease (IBD) in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
Also provided is a method of treating diarrhea in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (I-
C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing.
As microvascular complications can inhibit uniform delivery of anesthetics, in some embodiments the compounds herein may be used in methods of distributing local, regional, or systemic delivery of anesthetics and improve their effects. As such, also provided is a method of enhancing the delivery of anesthetics to a human experiencing microvascular complications, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically effective amount of an anesthetic. In some embodiments, the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt of any of the foregoing, is administered to the human prior to the administration of anesthetic.
Pharmaceutical Compositions
Also provided herein is a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal of any of the foregoing, and a pharmaceutically acceptable carrier or excipient. Additional pharmaceutical compositions comprise, respectively, a pharmaceutically effective amount of a compound selected from those of Formulas (I'), (I"), (I'"), (1-1), (1-2), (1-3), (1-4), (1-5), (1-6), and (1-7), and each of the formulas from
(l-a)-(l-ll), as well as the specifically named compounds herein, and a pharmaceutically acceptable carrier or excipient.
Further provided herein is the use of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (I-
E), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal of any of the foregoing, in the preparation of a medicament. Additional uses include the compounds that comprise, respectively, a pharmaceutically effective amount of a compound selected from those of Formulas (I'), (I"), (I'"), (1-1), (1-2), (1-3), (1-4), (1-5), (1-6), and (1-7), and each of the formulas from
(l-a)-(l-ll), as well as the specifically named compounds herein, for use in the preparation of a medicament.
The term "carrier" refers to an excipient or vehicle that includes without limitation diluents, disintegrants, precipitation inhibitors, surfactants, glidants, binders, lubricants, and the like with which the compound is administered. Pharmaceutically acceptable carriers are generally described herein and also in "Remington's Pharmaceutical Sciences" by E. W. Martin. Examples of carriers include, but are not limited to, aluminum monostearate, aluminum stearate, carboxymethylcellulose, carboxymethylcellulose sodium, crospovidone, glyceryl isostearate, glyceryl monostearate, hydroxyethyl cellulose, hydroxyethyl cellulose, hydroxymethyl cellulose, hydroxyoctacosanyl hydroxystearate, hydroxypropyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, lactose monohydrate, magnesium stearate, mannitol, microcrystalline cellulose, poloxamer 124, poloxamer 181, poloxamer 182, poloxamer 188, poloxamer 237, poloxamer 407, povidone, silicon dioxide, colloidal silicon dioxide, silicone, silicone adhesive 4102, and silicone emulsion. It should be understood, however, that the carriers selected for the pharmaceutical compositions, and the amounts of such carriers in the composition, may vary depending on the method of formulation (e.g., dry granulation formulation, solid dispersion formulation).
Some examples of suitable excipients in an oral formulation include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
As used herein, a "pharmaceutically acceptable excipient" is a pharmaceutically acceptable vehicle that includes, without limitation, any and all carriers, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
The pharmaceutical compositions may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, for example as described in those patents and patent applications incorporated by reference, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer.
One mode for administration is parenteral, particularly by injection. The forms in which the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of
Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles. Aqueous solutions in saline may also conventionally be used for injection. Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
Sterile injectable solutions are prepared by incorporating a compound according to the present disclosure in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. In some embodiments, for parenteral administration, sterile injectable solutions are prepared containing a therapeutically effective amount, e.g., 0.1 to
1000 mg, of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof. It will be understood, however, that the amount of the compound actually administered usually will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual subject, the severity of the subject's symptoms, and the like. Oral administration is another route for administration of the compound of Formula (l-A),
(l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co- crystal thereof. Administration may be via capsule or enteric coated tablets, or the like. In making the pharmaceutical compositions that include the compound of Formula (l-A), (l-B), (l-C), (l-D), or
(l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, the active ingredient is usually diluted by an excipient and/or enclosed within such a carrier that can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be in the form of a solid, semi-solid, or liquid material (as above), which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders.
In some embodiments, for parenteral administration, each dosage unit contains from 0.1 mg to 1 g, 0.1 mg to 700 mg, or 0.1 mg to 100 mg of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof.
In some embodiments, a therapeutically effective amount or a pharmaceutically effective amount of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of
Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt thereof, comprises from about 0.1 mg to about 500 mg per dose, given once or twice daily. In some embodiments, the individual dose is selected from 1 mg, 5 mg,
10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 75 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, and 500 mg per dose.
For any of the dosage units as described herein, it will be understood, however, that the amount of the compound actually administered usually will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual subject, the severity of the subject's symptoms, and the like. Articles of Manufacture and Kits
Compositions (including, for example, formulations and unit dosages) comprising the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula
(l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, can be prepared and placed in an appropriate container, and labeled for treatment of an indicated condition. Accordingly, provided is also an article of manufacture, such as a container comprising a unit dosage form of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula
(l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, and a label containing instructions for use of the compounds. In some embodiments, the article of manufacture is a container comprising a unit dosage form of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), ), a compound of any subformula of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), any of the specifically named compounds herein, or a pharmaceutically acceptable salt or co-crystal thereof, and at least one pharmaceutically acceptable vehicle. The article of manufacture may be a bottle, vial, ampoule, single-use disposable applicator, or the like, containing the pharmaceutical composition provided in the present disclosure. The container may be formed from a variety of materials, such as glass or plastic and in one aspect also contains a label on, or associated with, the container which indicates directions for use in the treatment of cancer or inflammatory conditions. It should be understood that the active ingredient may be packaged in any material capable of improving chemical and physical stability, such as an aluminum foil bag. In some embodiments, diseases or conditions indicated on the label can include, for example, treatment of cancer.
Definitions
Terms such as "microvascular tone", "microcirculatory tone", "microcirculatory vascular tone", and "arteriolar tone" refer to the degree of constriction experienced by a microcirculatory blood vessel relative to its maximally dilated state. Microcirculation may be anatomically defined as blood vessels having a diameter of between about 250 μ tom about 100 , μ pmarticularly including those between about 200 μm to about 150 μ,m and includes arterioles, capillaries, and venules (post-capillary venules). Collectively, these vessels may be referred to as "microvessels",
"microcirculatory vessels", and the like. In embodiments herein, it is a goal to provide compounds, compositions, and methods for relaxing microvessels in a subject, reducing vascular resistance and protecting the microvessels, themselves, and reducing or elimination end-organ damage associated with microvascular resistance, particularly that seen in the eyes, kidneys, and heart, as well as damage to nerve sheaths associated with microvessels. In some embodiments, the microcirculation is defined as blood vessels <200 μm in diameter. In the human body approximately 90% of the volume of blood resides in these vessels that include arterioles, capillary and venules. Arterioles range in diameter of approximately from 50-200 μm. Some venules contain vascular smooth muscle, and some capillaries are surrounded by pericytes, both of which are contractile cells that allow these vessels to constrict and relax, thus allowing more or less blood and oxygen to be delivered to cells, and cell waste removed. The size of these arterioles is tightly regulated to meet the oxygen demand of the cells they supply.
"Coronary microvascular disease" is heart disease that affects the walls and inner lining of tiny blood vessels branching from larger coronary arteries. Coronary microvascular disease may also be referred to as "Cardiac Syndrome X" or "nonobstructive coronary heart disease". In the heart or elsewhere it may also be referred to as "small artery disease", "small vessel disease", or
"arteriolosclerosis."
Such diseases of the small vessels are often seen in primary hypertension, stroke, and hypertensive renal disease. Manifestations as kidney lesions typically involve afferent arterioles and the interlobular artery and may involve intimal thickening, vascular smooth muscle cell proliferation, and extracellular matrix deposition, increasing the media-to-lumen ration. Areas of vascular smooth muscle cells may then be replaced by fibrosis and cell loss.
"Microvascular angina" refers to a form of chest pain due to abnormalities in cardiac microvessels, including, but not limited to, faulty relaxation of or spasms therein.
A "heterocycle" or "heterocyclic group" herein refers to a chemical ring containing carbon atoms and at least one ring heteroatom selected from O, S, and N, including saturated, unsaturated, partially saturated, and aromatic rings.
Examples of 3-membered heterocycles, such as seen in the definitions of R3 and R4 herein, include by way of example and not limitation aziridinyl, azirinyl, oxiranyl, and thiiranyl groups.
Examples of 4-membered heterocycles, such as seen in the definitions of R3 and R4 herein, include by way of example and not limitation azetidinyl, dihydro-1λ4-azetyl, azetyl, 1,3- diazetidinyl, and oxetanyl groups. Examples of 5-membered heterocycles, such as seen in the definitions of R3 and R4 herein, include by way of example and not limitation thiazolyl, tetrahydrothiophenyl, sulfur oxidized tetrahydrothiophenyl, furanyl, thienyl, pyrrolyl, dihydropyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, pyrrolidinyl, 2-pyrrolidonyl, dihydropyrrolidinyl, pyrrolinyl, tetrahydrofuranyl, triazinyl, thienyl, 2H- pyrrolyl, isothiazolyl, isoxazolyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, and oxazolidinyl groups.
Examples of 6-membered heterocycles, such as seen in the definitions of R3 and R4 herein, include by way of example and not limitation pyridyl, dihydroypyridyl, tetrahydropyridyl
(piperidyl), pyrimidinyl, piperidinyl, thiadiazinyl, thiazinyl, 2H,6H-1, 5, 2-dithiazinyl, pyranyl, pyrazinyl, pyridazinyl, piperazinyl, thienyl, thiopyran, dithanyl, morpholinyl, thiomorpholinyl, groups
Examples of 7-membered heterocycles herein, include by way of example and not limitation borepinyl, azepanyl, azepinyl, oxepanyl, oxepinyl, theipinyl, thiepanyl, diazepanyl, diazepinyl, and thiazepinyl groups.
Examples of 8-membered heterocycles herein, include by way of example and not limitation azocinyl, azocinyl, oxocanyl, oxocinyl, thiocanyl, and thiocinyl groups.
Examples of 9-membered heterocycles herein, include by way of example and not limitation monocyclic heterocycles such as azonanyl, azoninyl, oxonanyl, oxoninyl, thionanyl, and thioninyl groups and fused heterocycles, such as indolyl, indolinyl, isoindolyl, indolizinyl, indazolyl, azaindolyl, benzimidazolyl, azaindazolyl, pyrazolopyrimidinyl, purinyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, benzo[d]isoxazolyl, benzo[d]isothiazolyl, benzo[d]oxazolyl, benzo[c][l,2,5]thiadiazolyl, benzo[d]thiazolyl, benzisothiazolyl, adeninyl, and guaninyl groups.
Examples of 10-membered heterocycles herein, include by way of example and not limitation decahydroisoquinolinyl, decahydroquinolinyl, tetrahydroquinolinyl, quinolinyl, dihydroquinolinyl, isoquinolinyl, quinoxalinyl, quinolizinyl, phthalazinyl, quinazolinyl, cinnolinyl, chromenyl, isochromenyl, naphthyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[4,3-d]pyrimidinyl, pyrido[3,4-b]pyrazinyl, pyrido[2,3-b]pyrazinyl, pteridinyl, benzo[e][l,2]oxazinyl, benzo[e][l,3]oxazinyl, benzo[b][l,4]oxazinyl, quinolinonyl, thienoppyrimidinyl (including thieno[3,2-d]pyrimidinyl), and isoquinolinonyl groups.
It is understood that the general terms for the heterocycles referenced herein include each isomeric form of the heterocycle, such as the term "dithianyl" including 1,2 dithianyl, 1,3- dithianyl, and 1,4-dithianyl groups, the term "thiadiazinyl" including 1,2,5 thiadiazineyl and 1,3,4- thiadiainyl groups, the term "azaindolyl" including 4-azaindolyl, 5-azaindolyl, 6-azaindolyl, and 7- azaindolyl groups, and "benzothiophenyl" including benzo[b]thiophenyl and benzo[c]thiophenyl groups.
Similarly, general heterocycle names include each variance in one or more points of unsaturation. For instance, the term "dihydropyrrolyl" refers to "2,3-dihydro-lH-pyrrolyl" and
"2,5-dihydro-lH-pyrrolyl" groups.
The term "alkyl" refers to a straight or branched hydrocarbon. For example, an alkyl group can have a specified number of chain carbons, such as 1 to 6 carbon atoms (i.e., C1-C6 alkyl or C1-6 alkyl). Examples of suitable alkyl groups include, but are not limited to, methyl (Me, -CH3), ethyl
(Et, -CH2CH3), 1-propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, --CH(CH3)2), 1-butyl (n-
Bu, n-butyl, --CH2CH2CH2CH3), 2-methyl-l-propyl (i-Bu, i-butyl, --CH2CH(CH3)2), 2-butyl (s-Bu, s- butyl, --CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, --C(CH3)3), 1-pentyl (n-pentyl, -
CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (— CH(CH2CH3)2), 2-methyl-2-butyl (-
C(CH3)2CH2CH3), 3-methyl-2-butyl (— CH(CH3)CH(CH3)2), 3-methyl-l-butyl (-CH2CH2CH(CH3)2), 2- methyl-l-butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (-
CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-
C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-
CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-
CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (— C(CH3)2CH(CH3)2), and 3,3-dimethyl-2-butyl (-
CH(CH3)C(CH3)3.
The term "alkoxy" refers to a group having the formula "-O-alkyl," in which an alkyl group, as defined above, is attached to the parent molecule via an oxygen atom. The alkyl portion of an alkoxy group can have a specified number of carbon chain atoms, such as 1 to 6 carbon atoms
(i.e., C1-C6 alkoxy or C1-6 alkoxy). Examples of suitable alkoxy groups include, but are not limited to, methoxy (-O-CH3 or --OMe), ethoxy (-OCH2CH3 or --OEt), t-butoxy (--O--C(CH3)3 or --OtBu) and the like.
The term "carbocycle" or "carbocyclic group" refers to a chemical ring containing only carbon atoms, including saturated, unsaturated, partially saturated, and aromatic rings.
The term "cycloalkyl" refers to a saturated ring having 3 to 6 carbon atoms as a monocycle, including cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl groups. The term "halogen" refers to an atom selected from the group of elements chlorine, fluorine, bromine, and iodine.
The term "oxo" refers to double-bonded oxygen "=O".
The terms "therapeutically effective amount" and "pharmaceutically effective amount" refer to an amount that is sufficient to effect treatment, as defined below, when administered to a subject (e.g., a mammal, such as a human) in need of such treatment. The therapeutically or pharmaceutically effective amount will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
For example, a "therapeutically effective amount" or a "pharmaceutically effective amount" of a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt or cocrystal thereof, is an amount sufficient to antagonize GPR39 expression or activity, and thereby treat a subject (e.g., a human) suffering an indication, or to ameliorate or alleviate the existing symptoms of the indication. For example, a therapeutically or pharmaceutically effective amount may be an amount sufficient to decrease a symptom of a disease or condition responsive to inhibition of GPR39 activity.
"Treatment" or "treating" is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired clinical results may include one or more of the following: (i) inhibiting the disease or condition (e.g., decreasing one or more symptoms resulting from the disease or condition, and/or diminishing the extent of the disease or condition); (ii) slowing or arresting the development of one or more clinical symptoms associated with the disease or condition (e.g., stabilizing the disease or condition, preventing or delaying the worsening or progression of the disease or condition, and/or preventing or delaying the spread
(e.g., metastasis) of the disease or condition); and/or (iii) relieving the disease, that is, causing the regression of clinical symptoms (e.g., ameliorating the disease state, providing partial or total remission of the disease or condition, enhancing effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival).
The terms "inhibiting" or "inhibition" indicates a decrease, such as a significant decrease, in the baseline activity of a biological activity or process. "Inhibition of GPR39 activity" refers to a decrease in GPR39 activity as a direct or indirect response to the presence of a compound of
Formula (l-A), (l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt or co-crystal thereof, relative to the activity of GPR39 in the absence of such compound or a pharmaceutically acceptable salt or co-crystal thereof. The decrease in activity may be due to the direct interaction of the compound with GPR39, or due to the interaction of the compound(s) described herein with one or more other factors that in turn affect GPR39 expression and/or activity. For example, the presence of the compound(s) may decrease GPR39 activity by directly binding to the GPR39, by causing (directly or indirectly) another factor to decrease GPR39 expression or activity, or by
(directly or indirectly) decreasing the amount of GPR39 protein present in the cell or organism. In some embodiments, the inhibition of GPR39 activity may be compared in the same subject prior to treatment, or other subjects not receiving the treatment. The term "inhibitor" is understood to refer to a compound or agent that, upon administration to a human in need thereof at a pharmaceutically or therapeutically effective dose, provides the inhibition activity desired.
"Delaying" the development of a disease or condition means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease or condition. This delay can be of varying lengths of time, depending on the history of the disease or condition, and/or subject being treated. A method that "delays" development of a disease or condition is a method that reduces probability of disease or condition development in a given time frame and/or reduces the extent of the disease or condition in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects. Disease or condition development can be detectable using standard methods, such as routine physical exams, mammography, imaging, or biopsy. Development may also refer to disease or condition progression that may be initially undetectable and includes occurrence, recurrence, and onset.
As used herein, the singular terms "a," "an," and "the" include plural referents unless context clearly indicates otherwise. Similarly, the word "or" is intended to include "and" unless the context clearly indicates otherwise. Also, as used herein, the term "comprises" means
"includes." Hence "comprising A or B" means including A, B, or A and B.
Numerical values in the specification and claims of this application should be understood to include numerical values which are the same when reduced to the same number of significant figures and numerical values which differ from the stated value by less than the experimental error of conventional measurement technique of the type described in the present application to determine the value. All ranges disclosed and/or claimed herein are inclusive of the recited endpoint and independently combinable (for example, the ranges of "from 2 to 10" and "2-10" are inclusive of the endpoints, 2 and 10, and all the intermediate values 3, 4, 5, 6, 7, 8, and 9).
By "significant" is meant any detectable change that is statistically significant in a standard parametric test of statistical significance such as Student's T-test, where p<0.05.
"Pharmaceutically acceptable salts" include, for example, salts with inorganic acids and salts with an organic acid. Examples of salts may include hydrochloride, phosphate, diphosphate, hydrobromide, sulfate, sulfinate, nitrate, malate, maleate, fumarate, tartrate, succinate, citrate, acetate, lactate, methanesulfonate (mesylate), benzenesuflonate (besylate), p-toluenesulfonate
(tosylate), 2-hydroxyethylsulfonate, benzoate, salicylate, stearate, and alkanoate (such as acetate,
HOOC--(CH2)n--COOH where n is 0-4). In addition, if the compounds described herein are obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt.
Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare nontoxic pharmaceutically acceptable addition salts.
The term "crystal forms" and related terms herein refer to the various crystalline modifications of a given substance, including, but not limited to, polymorphs, solvates, hydrates, co-crystals, and other molecular complexes, as well as salts, solvates of salts, hydrates of salts, other molecular complexes of salts, and polymorphs thereof. Crystal forms of a substance can be obtained by a number of methods, as known in the art. Such methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystallization on surfaces or templates, such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal countermolecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding and solvent-drop grinding.
The term "co-crystal" or "co-crystal salt" as used herein means a crystalline material composed of two or more unique solids at room temperature, each of which has distinctive physical characteristics such as structure, melting point, and heats of fusion, hygroscopicity, solubility, and stability. A co-crystal or a co-crystal salt can be produced according to a per se known co-crystallization method. The terms co-crystal (or cocrystal) or co-crystal salt also refer to a multicomponent system in which there exists a host API (active pharmaceutical ingredient) molecule or molecules, such as a compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), and a guest (or co-former) molecule or molecules. In particular embodiments the pharmaceutically acceptable co-crystal of the compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E)with a co-former molecule is in a crystalline form selected from a malonic acid co-crystal, a succinic acid co-crystal, a decanoic acid co-crystal, a salicylic acid co-crystal, a vanillic acid co-crystal, a maltol co-crystal, or a glycolic acid co-crystal. Co-crystals may have improved properties as compared to the parent form (i.e., the free molecule, zwitter ion, etc.) or a salt of the parent compound. Improved properties can include increased solubility, increased dissolution, increased bioavailability, increased dose response, decreased hygroscopicity, a crystalline form of a normally amorphous compound, a crystalline form of a difficult to salt or unsalable compound, decreased form diversity, more desired morphology, and the like.
As used herein, the terms "isotope" and "isotopic" in reference to a compound as disclosed herein means that one or more atoms of the compound is replaced with an isotope of such one or more atoms. An "isotope" refers to any of two or more forms of a chemical element, having the same number of protons in the nucleus, but having different numbers of neutrons in the nucleus. For example, an isotopic compound includes a compound in which one or more hydrogen atoms (H) has been replaced with one or more deuterium atoms (D). In this example, deuterium is an isotope of hydrogen, and replacing a hydrogen atom with deuterium (at one or more positions) renders the resulting compound an isotopic compound. For example, and in reference to Formula (l-A), (l-B), (l-C), (l-D), or (l-E), replacing the two methyl groups of the isopropyl moiety (-CH(CH3)2) with fully deuterated methyl groups (-CH(CD3)2) would be an isotopic compound of Formula (l-A), (l-B), (l-C), (l-D), or (l-E). In addition to replacing hydrogen with deuterium, other stable (non-radioactive) isotope substitutions include replacing carbon 12 with carbon 13, while unstable (radioactive) isotopes include replacing hydrogen with tritium, replacing carbon 12 with carbon 14, replacing iodine 127 with iodine 123 or iodine 125, and the like.
Accordingly, all reference herein to isotopic compounds of Formula (l-A), (l-B), (l-C), (l-D), or (l-E), as well as all reference to the various embodiments thereof, refers to a compound having one or more isotopic substitutions, including (but not limited to) substitutions of one or more hydrogen atoms with one or more deuterium atoms and any occurrence(s) in the compound. To this end, the isotopic compounds disclosed herein provide improved advantages relative to their non- isotopic forms. To this end, isotopic modification provides a means of improving existing drugs and/or as a tool in the design of new drugs. For example, isotopic drug design has proven successful in the context of the deuterium (D) kinetic isotope effect. Due to the twofold higher mass of D compared with H, the C-D bond is much more resistant toward oxidative processes (such as its ability to be catalyzed by CYP450 or by other enzymes involved in metabolism), while retaining very similar steric properties. Therefore, H-D isosteric replacement usually retains the pharmacodynamics of the compound, while improving its pharmacokinetics with a repercussion on half-life and/or of area under the curve values and, ultimately, on dose and/or dosing regimen.
For example, drug exposure may be enhanced with isotopic modification, and/or a decrease of clearance. Such benefits are provided to the compounds disclosed hereby by way their isotopic derivation.
Terms such as "subject" and "patient" refer to an animal, such as a mammal, that has been or will be the object of treatment, observation or experiment. The methods described herein may be useful in both human therapy and veterinary applications. In some embodiments, the subject is a mammal; in some embodiments the subject is human; and in some embodiments the subject is chosen from cats and dogs. "Subject in need thereof" or "human in need thereof" refers to a subject, such as a human, who may have or is suspected to have diseases or conditions that would benefit from certain treatment; for example treatment with a compound of Formula (l-A),
(l-B), (l-C), (l-D), or (l-E), or a pharmaceutically acceptable salt or co-crystal thereof, as described herein. This includes a subject who may be determined to be at risk of or susceptible to such diseases or conditions, such that treatment would prevent the disease or condition from developing.
The terms "prediabetes" or "prediabetic condition" refers to a condition in which a subject's blood sugar levels are not high enough to be considered diabetic but may be a precursor to Type 2 diabetes. A prediabetic condition may be defined in subjects having a fasting blood glucose level of 100 mg/dl or more, but less than 126 mg/dl (the level which is diagnostic for diabetes). Hemoglobin A1c (HbA1c) levels are another laboratory test for diabetes. HbA1c levels of 6.5% or greater are characteristic of diabetes, while levels from 5.7% to 6.4% suggest prediabetes.
Examples
Measurement of intracellular calcium response in PC3 cells, expressing the endogenous human
GPR39 receptor
Human prostate adenocarcinoma (PC3) cells endogenously expressing human GPR39 receptor were seeded into black walls clear-base 384-well plates at a density of 7,500 cells per well in DM EM (Low Glucose) supplemented with 10% heat-inactivated foetal bovine serum and 1% of
Pen/Strep and grown overnight at 37 °C, 5% CO2. After washing with assay buffer (20 mM HEPES,
137 mM NaCI, 5.4 mM KCI, 10 mM glucose, 0.8 mM MgSO4, 1.3 mM CaCI2, 0.3 mM Na2HPO4, 0.4 mM KH2PO44.2 mM NaHCO3, pH 7.4), cells were incubated with Calcium-6 probe (Molecular
Devices) at 37°C for 2h and then equilibrated at RT for 30 minutes on metal block. The fluorescence was measured in real time by Fluorometric Imaging Plate Reader (FLIPR TETRA,
Molecular Devices) (excitation wavelength at 470-495 nm, emission wavelength at 515-575 nm).
Compounds of invention were serially diluted in neat DMSO at 1000 fold concentrations and plated into 384-well plates and then 200-fold diluted in assay buffer containing 150 μM ZnCI2 (30 μM final concentration) and 1.1% DMSO (0.32% final concentration).
A dual addition FLIPR protocol was used which included a first addition of compounds 200-fold diluted in Assay Buffer containing 150 μM ZnCI2 and 1.1% DMSO and then after 10 minutes a second addition of a submaximal concentration of the hGPR39 receptor agonist, C3 (Tocris, TC-G
1008) at EC90 (concentration producing 90% of the maximal response) value of 1.5 μM.
Fluorescence was monitored throughout the run assay. The peak of Ca2+ stimulation (baseline subtracted) was expressed as percentage of the maximal inhibition of the EC90 C3 response and was plotted versus the concentration of test compound. Curve fitting and IC50 estimations are carried out using a four-parameter logistic model using XLfit Software.
The potency of compounds was reported as functional pKi (-LoglOKi) obtained using the modified
Cheng-Prusoff relationship (Cheng, Y., Prusoff, W.H. 1973.) f Ki =IC50/(l+[L]/EC50) where IC50 is the concentration of antagonist required for 50% inhibition of the maximum response, [L] is the concentration of the agonist used (EC90) and EC50 is the concentration of agonist need to induced 50% of the maximum response (obtained in each experimental plate). fpKi data is shown in Table 1 below. An fpKi less than 5.2 is reported as "+/-". An fpKi ranging from 5.2 to 5.9 is reported as "+". An fpKi from 6.0 to 6.5 is reported as "++". An fpKi from 6.6 to
7.2 is reported as "+++". An fpKi equal to or greater than 7.3 is reported as "++++". Table 1
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Synthesis
The compounds described herein may be prepared by methods known in the art and are exemplified by the following non-limiting descriptions.
Unless otherwise stated, all reactions are typically performed under inert atmosphere (for example under Nitrogen). The following abbreviations are used in the text:
EtOAc, AcOEt, EA = ethyl acetate
Et2O = diethyl ether
CH3CN = acetonitrile
MeOH = methanol
THF = tetra hydrofuran r.t. (RT) = room temperature
DMSO = dimethyl sulfoxide
DMF = N,N'-dimethylformamide
DCM = dichloromethane
EtOH = ethanol
RP = reverse phase
FA = formic acid
Cy, cHex = cyclohexane
TEA = triethylamine
DIPEA = N, N- Diisopropylethylamine
BOC2O = Di-tert-butyl dicarbonate
TFA = trifluoroacetic acid
HATU = l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate
AcOH = acetic acid
SCX Cartridge = Strong Cation Exchange Cartridge
FC = Flash chromatography s.s. = saturated solution
ON = overnight
Scheme 1
Figure imgf000070_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Enamine) by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. CH3CN, typically at
80° C. The reaction takes about 1 hour to complete.
Step 2
Compound of formula III may be obtained by nitro reduction of compound of formula II under catalytic hydrogenation conditions, using for example Pd/C in a suitable solvent such as acetic acid, typically at room temperature. The reaction takes about 4 hours to complete
Step 3
Compound of formula IV may be obtained by coupling reaction between compound of formula III and a proper commercially available acyl chloride, e.g. cyclopropanecarbonyl chloride, in presence of an organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 12 hours to complete. Step 4
Compound of formula V may be obtained from compound of formula IV by hydrolysis, e.g. using
LiOH, in a suitable mixture of solvents such as MeOH/THF/H2O, typically at 50 °C. The reaction takes about 36 hours to complete.
Step 5
Compound of formula VI may be obtained from compound of formula V by coupling with the proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
Scheme 1, Cpd II Preparation: methyl 4-nitro-2-pyrrolidin-l-ylbenzoate
Figure imgf000071_0001
To a solution of Methyl 2-Fluoro-4-nitrobenzoato (5.0 g, 25.11 mmol) in MeCN (50 mL), pyrrolidine
(5.36 g, 75.32 mmol) was added and the reaction was stirred at 80 °C for 1 h. After this time the reaction was cooled down to RT and then concentrated under vacuum. The residue was taken up with water and filtered. The solid was dried under vacuum affording the product of formula methyl
4-nitro-2-pyrrolidin-l-ylbenzoate which was used in the next step without further purification.
Yield: 6 g
1H NMR (400 MHz, DMSO-d6) δ 7.64 (d, 1H), 7.49 (d, 1H), 7.44 (dd, 1H), 3.86 (s, 3H), 3.26 - 3.20 (m,
4H), 1.95 - 1.90 (m, 4H); LC-MS: 251.05 (MH+).
Scheme 1, Cpd III Preparation: methyl 4-amino-2-pyrrolidin-l-ylbenzoate
Figure imgf000071_0002
To a solution of methyl 4-nitro-2-pyrrolidin-l-ylbenzoate (4.75 g, 18.98 mmol), in Acetic acid (190 mL) Pd-C 10% (2.02 g, 1.9 mmol) was added. The reaction was stirred for 4 hours under Hg atmosphere at room temperature. After this time the mixture was filtered through a celite pad, washed with MeOH and the solvent was evaporated under vacuum. The residue was taken up with a s. s. of NaHCO3 and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula methyl 4-amino-2-pyrrolidin-l-yl benzoate, which was used in the next step without further purification.
Yield: 3.95 g
1H NMR (400 MHz, DMSO-d6) δ 7.29 (d, 1H), 6.02 - 5.88 (m, 2H), 5.47 (s, 2H), 3.67 (s, 3H), 3.16 -
2.99 (m, 4H), 2.00 - 1.71 (m, 4H); LC-MS: m/z 221.39 (MH+).
Scheme 1, Cpd IV Preparation: methyl 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate
Figure imgf000072_0001
A solution of methyl 4-amino-2-pyrrolidin-l-ylbenzoate (5.75 g, 26.1 mmol) and N,N-
Diisopropylethylamine (9.09 mL, 52.21 mmol) in dry DCM (145.57 mL) was cooled to 0 °C and stirred for 10 min.. After this time cyclopropanecarbonyl chloride (3.08 mL, 33.94 mmol) was added dropwise and the reaction mixture was allowed to reach room temperature and stirred overnight.
The day after the reaction was diluted with DCM and washed with a saturated solution of NaHCO3.
The organic phase was washed with brine, dried over a phase separator and the solvent was evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from cHex/ AcOEt 9:1 to cHex/ AcOEt 9:1) to afford the product of formula methyl 4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate.
Yield: 5.6 g
1H NMR (400 MHz, DMSO-d6) δ 10.22 (s, 1H), 7.41 (d, 1H), 7.22 (d, 1H), 6.92 (dd, 1H), 3.75 (s, 3H),
3.11 (q, 4H), 1.92 - 1.83 (m, 4H), 1.83 - 1.75 (m, 1H), 0.83 - 0.76 (m, 4H); LC-MS: m/z 289.12 (MH+).
Scheme 1, Cpd V Preparation: 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoic acid
Figure imgf000073_0002
To a solution of methyl 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate (5.0 g, 17.34 mmol) in THF (150 mL) / MeOH (20 mL), a 6N solution of NaOH (5.78 mL, 34.68 mmol) was added.
The reaction was stirred at 50 °C overnight.
The day after further sodium hydroxide solution 6 N (5.78 mL, 34.68 mmol) was added followed by
10 mL of MeOH and the reaction was stirred at 50 °C for 1 day. After this time the reaction was cooled down to RT and concentrated under vacuum. The aqueous solution was washed with DCM
(3 times), acidified with HCI 12 N until pH = 1 and extracted with AcOEt 5 times. The combined organic fractions were washed with Brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was taken up with Et2O and filtered, the solid was washed with Et2O and dried under vacuum affording the product of formula 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoic acid, which was used in the next step without further purification.
Yield: 2.5 g
1H NMR (400 MHz, DMSO-d6) δ 13.10 (s, 1H), 10.25 (s, 1H), 7.52 (d, 1H), 7.34 (d, 1H), 6.99 (dd, 1H),
3.21 - 3.04 (m, 4H), 2.01 - 1.78 (m, 4H), 1.79 (tt, 1H), 0.86 - 0.73 (m, 4H); LC-MS: m/z 275.08 (MH+).
Preparation: N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000073_0001
Example 1
To a solution of 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-yl benzoic acid (46.58 mg, 0.170 mmol) and (HATU) [dimethylamino(3-triazolo[4,5-b]pyridinyloxy)methylidene]- dimethylammonium hexafluorophosphate (96.85 mg, 0.250 mmol) and N,N-Diisopropylethylamine
(0.09 mL, 0.510 mmol) in DMF (2 mL) a solution 2-phenylpyrrolidine (25.0 mg, 0.170 mmol) in DMF
(ImL) was added. The reaction mixture was stirred at RT/ON. After completion of the reaction the mixture was diluted with a s. s. of NaHCO3and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from cHex/ AcOEt 8:2 to cHex/ AcOEt 2:8) affording the product of formula N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide (see Example 1).
Yield: 34.2 mg
NMR (400 MHz, DMSO-d6) δ 10.14 - 9.87 (m, 1H), 7.38 - 7.27 (m, 3H), 7.26 - 7.12 (m, 2H), 7.09 -
6.92 (m, 3H), 5.17 - 4.78 (m, 1H), 3.61 (d, 2H), 3.08 (d, 4H), 2.39 - 2.17 (m, 1H), 2.04 - 1.62 (m, 8H),
0.84 - 0.62 (m, 4H); LC-MS: m/z 404.3 (MH+).
Preparation: N-[4-(3-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000074_0001
Example 2
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-phenylpyrrolidine was used instead of 2- phenylpyrrolidine. The title compound was obtained in a yield of 38 % (26 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.08 (d, 1H), 7.39 - 7.18 (m, 5H), 7.10 - 6.97 (m, 2H), 6.96 - 6.85
(m, 1H), 3.99 - 3.65 (m, 2H), 3.38 (d, 3H), 3.26 - 2.99 (m, 4H), 2.25 (d, 1H), 2.08 - 1.70 (m, 6H), 0.77
(dtd, 4H); LC-MS: m/z 404.3 (MH+).
Preparation: N-[4-(2-oxa-7-azaspiro[3.4]octane-7-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000074_0002
Example 3
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 2-oxa-6-azaspiro[3.4]octane was used instead of 2- phenylpyrrolidine. The title compound was obtained in a yield of 61 % (40 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 7.10 - 7.02 (m, 1H), 6.99 - 6.86 (m, 2H), 4.57 (d, 1H),
4.49 - 4.38 (m, 3H), 3.65 (s, 1H), 3.41 (s, 2H), 3.11 (d, 5H), 2.12 (dt, 2H), 1.89 - 1.81 (m, 4H), 1.77 (qd,
1H), 0.77 (dt, 4H); LC-MS: m/z 370.2 (MH+).
Preparation: N-[4-[(trans)-2,6-dimethylmorpholine-4-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000075_0001
Example 4
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein trans-2,6-dimethylmorpholine was used instead of 2- phenylpyrrolidine. The title compound was obtained in a yield of 50 % (33 mg) as a racemic mixture.
1H NMR (400 MHz, DMSO-d6) δ ppm 10.09 (s, 1H), 7.06 (s, 1H), 6.99 - 6.88 (m, 2H), 4.11 - 3.67 (m,
3H), 3.41 - 2.72 (m, 7H), 1.95 - 1.80 (m, 4H), 1.80 - 1.70 (m, 1H), 1.32 - 1.09 (m, 3H), 0.99 (dd, 3H),
0.82 - 0.69 (m, 4H); LC-MS: m/z 372.23 (MH+).
Preparation: N-[4-(2,6-dimethyl-l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000075_0002
Example 5
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 2,6-dimethyl-l,4-thiazinane 1,1-dioxide hydrochloride was used instead of 2-phenylpyrrolidine. The title compound was obtained in a yield of 44 % (34 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.13 (d, 1H), 7.28 - 6.72 (m, 3H), 4.79 (d, 1H), 3.83 (dd, 1H), 3.48 -
3.38 (m, 1H), 3.25 - 2.76 (m, 7H), 1.97 - 1.65 (m, 5H), 1.25 (t, 3H), 1.06 (dd, 3H), 0.81 - 0.74 (m, 4H);
LC-MS: m/z 420.2 (MH+).
Preparation: N-[4-(8-oxa-3-azabicyclo[3.2.1]octane-3-carbonyl)-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide
Figure imgf000076_0001
Example 6
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 8-oxa-3-azabicyclo[3.2.1]octane hydrochloride was used instead of 2-phenylpyrrolidine. The title compound was obtained in a yield of 44 % (34 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.09 (s, 1H), 7.06 (dd, 1H), 7.00 - 6.82 (m, 2H), 4.47 - 4.00 (m, 3H),
3.24 - 2.82 (m, 7H), 1.98 - 1.44 (m, 9H), 0.77 (dt, 4H); LC-MS: m/z 370.2 (MH+).
Preparation: N-[4-(3-oxa-8-azabicyclo[3.2.1]octane-8-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000076_0002
Example 7
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-oxa-8-azabicyclo[3.2.1]octane hydrochloride was used instead of 2-phenylpyrrolidine. The title compound was obtained in a yield of 13 % (9 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.13 (s, 1H), 7.33 - 6.83 (m, 3H), 4.53 (s, 1H), 3.84 - 3.41 (m, 5H),
3.23 - 2.94 (m, 4H), 1.97 - 1.70 (m, 9H), 0.82 - 0.67 (m, 4H); LC-MS: m/z 370.2 (MH+). Preparation: N-[4-(6-oxa-3-azabicyclo[3.1.1]heptane-3-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000077_0002
Example 8
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 6-oxa-3-azabicyclo[3.1.1]heptane hydrochloride was used instead of 2-phenylpyrrolidine. The title compound was obtained in a yield of 75 % (49 mg).
1H NMR (400 MHz, DMF-d7) δ 10.07 (s, 1H), 7.07 (d, 1H), 7.01 - 6.85 (m, 2H), 4.53 (d, 2H), 3.84 (d,
1H), 3.61 - 3.44 (m, 2H), 3.26 (s, 2H), 3.24 - 2.93 (m, 5H), 1.94 - 1.56 (m, 5H), 0.77 (dd, 4H); LC-MS: m/z 356.2 (MH+)
Preparation: N-[4-(3-azabicyclo[3.1.1]heptane-3-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000077_0001
Example 9
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-oxa-8-azabicyclo[3.2.1]octane hydrochloride was used instead of 2-phenylpyrrolidine. The title compound was obtained in a yield of 78 % (50 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.06 (s, 1H), 7.05 (s, 1H), 6.93 (s, 2H), 3.73 - 3.34 (m, 4H), 3.29 -
3.01 (m, 6H), 2.39 - 2.32 (m, 1H), 2.17 - 2.04 (m, 2H), 1.97 - 1.68 (m, 4H), 1.33 - 1.13 (m, 2H), 0.78
(dd, 4H); LC-MS: m/z 354.2 (MH+)
Preparation: tert-butyl N-[l-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperidin-4- yl]-N-methylcarbamate
Figure imgf000078_0001
Example 10
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 4-N-BOC-4-N -methyl -aminopiperidine was used instead of 2-phenylpyrrolidine. The title compound was obtained in a quantitative yield (140 mg).
LC-MS: m/z 471.49 (MH+)
Preparation: tert-butyl N-[l-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperidin-4- yl)carbamate
Figure imgf000078_0002
Example 11
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein N-(4-piperidinyl)carbamic acid tert-butyl ester was used instead of 2-phenylpyrrolidine. The title compound was obtained in a quantitative yield (140 mg).
LC-MS: m/z 457.48 (MH+)
Preparation: tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate
Figure imgf000078_0003
Example 12 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-[2-(trifluoromethyl)phenyl]piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a yield of 19% (24mg).
LC-MS: m/z 587.30 (MH+).
Preparation: tert-butyl 3-(3-cyanophenyl)-4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoyl]piperazine-l-carboxylate
Figure imgf000079_0002
Example 13
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-cyanophenyl)piperazine-l-carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a yield of 47% (55mg).
LC-MS: m/z 544.27 (MH+).
Preparation: tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-(4,5- dimethyl-l,3-thiazol-2-yl)piperazine-l-carboxylate
Figure imgf000079_0001
Example 14
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(4,5-dimethyl-l,3-thiazol-2-yl)piperazine- 1-carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a yield of 60% (73 mg).
LC-MS: m/z 554.25 (MH+)
Preparation: tteerrtt--bbuuttyyll 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-(4- methylphenyl)piperazine-l-carboxylate
Figure imgf000080_0001
Example 15
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(4-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a yield of 70% (96 mg).
Preparation: tert-butyl 3-(3-chloro-5-methylphenyl)-4-[4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoyl]piperazine-l-carboxylate
Figure imgf000080_0002
Example 16
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-chloro-5-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a yield of 90% (130 mg).
LC-MS: m/z 567.29 (MH+). Preparation: tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-(3- methyl phenyl)piperazine-l-carboxylate
Figure imgf000081_0001
Example 17
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 1 N-[4-(2-phenylpyrrolidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-methylphenyl)piperazine-l- carboxylate was used instead of 2-phenylpyrrolidine. The title compound was obtained as a racemic mixture in a quantitative yield (150 mg).
LC-MS: m/z 533.33 (MH+).
Scheme 2
Figure imgf000081_0002
Step 1
Compound of formula II may be obtained via acyl chloride formation of compound of formula I
(commercially available from Enamine) by treatment with a suitable chlorinating agent, such as
SOCl2, typically at 90 °C, followed by coupling with l-Methyl-3-phenylpiperazine (commercially available from Enamine) in the presence of a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 12 hours to complete. Step 2
Compound of formula II may be obtained from compound of formula II by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes about 36 hours to complete.
Step 3
Compound of formula IV may be obtained from compound of formula III by Buchwald reaction with the proper heteroaryl aniline in the presence of a suitable palladium precatalyst, e.g. tBuBrettPhos Pd G3, and a suitable base, such as K3PO4. The reaction is carried out in a suitable solvent, e.g. 1- butanol, typically at 120 °C. The reaction takes from about 12 hours to about 24 hours to complete.
Step 4
Compound of formula V may be obtained via Suzuki coupling between compound of formula III and the appropriate commercially available boronic acid in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na2CO3, in a suitable solvent, 1,2- dimethoxyethane, typically at 95 °C. The reaction takes about 2 days to complete.
Scheme 2, Cpd II Preparation: (2-fluoro-4-iodophenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000082_0001
A mixture 2-fluoro-4-iodobenzoic acid (2.0 g, 7.52 mmol) and thionyl dichloride (10.97 mL, 150.37 mmol) was heated to reflux for 2.5 hours. After this time the reaction was cooled down to RT and then concentrated under vacuum. The mixture was stripped three times with cyclohexane in order to remove the excess of thionyl dichloride. The residue was suspended in dry DCM (40 mL) and triethylamine (3.14 mL, 22.56 mmol) was added followed by l-methyl-3-phenylpiperazine (1.33 g,
7.52 mmol). The reaction mixture was stirred overnight at RT. The day after, the mixture was diluted with DCM and washed with a s.s NaHCO3. The combined organic phases were dried over a phase separator and concentrated under vacuum affording the product of formula (2-fluoro-4- iodophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone as racemic mixture. The product was used in the next step without further purification.
Yield: 3.2 g
NMR (400 MHz, DMSO-d6) δ 7.75 (dd, 2H), 7.47 (d, 2H), 7.42 - 7.20 (m, 4H), 4.74 - 3.64 (m, 1H),
3.55 - 3.34 (m, 1H), 3.25 - 2.57 (m, 2H), 2.36 - 2.24 (m, 1H), 2.18 (d, 3H), 2.07 - 1.83 (m, 1H), 0.97 (t,
1H); LC-MS: m/z 425.05 (MH+).
Scheme 2, Cpd III Preparation: (4-iodo-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000083_0001
To a solution of (2-fluoro-4-iodophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone (2.79 g, 6.56 mmol) in DMSO (50 mL) pyrrolidine (1.64 mL, 19.69 mmol) was added and the reaction mixture was stirred ON at 120 °C. After 16 h further pyrrolidine (0.55 mL, 6.56 mmol) was added and the reaction was stirred for 1 day. The day after the reaction was cooled down to RT and diluted with water and the aqueous phase was extracted with AcOEt three times. The combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of
HCOOH to 95:5 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted 3 times with AcOEt.
The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula (4-iodo-2-pyrrolidin-l-ylphenyl)-(4- methyl-2-phenylpiperazin-l-yl)methanone.
Yield: 1.0 g
1H NMR (400 MHz, DMSO) δ 7.73 (d, 1H), 7.53 - 7.14 (m, 4H), 7.10 - 6.76 (m, 2H), 6.68 (d, 1H), 5.84
- 5.57 (m, 1H), 4.77 - 4.29 (m, 1H), 3.14 - 2.58 (m, 7H), 2.43 - 1.99 (m, 7H), 1.75 - 1.49 (m, 2H); LC-
MS: m/z 476.1 (MH+).
Preparation: [4-[(3-methyl-l,2-oxazol-5-yl)amino]-2-pyrrolidin-l-ylphenyl]-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000084_0002
Example 18
A mixture of (4-iodo-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone (150.0 mg, 0.320 mmol), 3-methyl-5-isoxazolamine (37.15 mg, 0.380 mmol), tripotassium phosphate
(66.98 mg, 0.320 mmol) and tBuBrettPhos Pd G3 (26.96 mg, 0.030 mmol) in 1-Butanol (1.2 mL) was degassed by N2-vacuum cycles. The reaction was stirred at 120 °C for 24 hours. After this time the reaction was cooled down to RT, diluted with MeOH and purified by SCX first washing with MeOH and then eluting with NH3 IN in MeOH. Basic fractions were collected and concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 95:5 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted 3 times with AcOEt. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula [4-[(3-methyl- l,2-oxazol-5-yl)amino]-2-pyrrolidin-l-ylphenyl]-(4-methyl-2-phenylpiperazin-l-yl)methanone as a racemic mixture.
Yield: 16 mg
NMR (400 MHz, DMSO-d6) δ 9.75 (d, 1H), 7.83 - 7.14 (m, 6H), 6.97 (ddd, 1H), 6.78 - 6.66 (m, 1H),
6.61 - 6.35 (m, 1H), 3.55 - 3.30 (m, 2H), 3.10 - 2.55 (m, 7H), 2.37 - 2.10 (m, 7H), 1.94 (s, 4H), 1.67 (d,
1H); LC-MS: m/z 446.48 (MH+).
Preparation: (4-methyl-2-phenylpiperazin-l-yl)-[2-pyrrolidin-l-yl-4-([l,2,4]triazolo[l,5-a]pyridin-2- ylamino)phenyl]methanone
Figure imgf000084_0001
Example 19 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 18 [4-[(3-methyl-l,2-oxazol-5-yl)amino]-2-pyrrolidin-l-ylphenyl]-(4-methyl-2- phenylpiperazin-l-yl)methanone wherein [l,2,4]triazolo[l,5-a]pyridin-2-amine was used instead of
3-methyl-5-isoxazolamine. The title compound was obtained in a yield of 16 % (25 mg).
1H NMR (500 MHz, DMSO-d6 ) δ ppm 9.52 (br d, 1H), 8.74 (d, 1H), 7.84 - 6.50 (m, 11H), 5.93 - 4.61
(m, 1H), 4.54 - 3.36 (m, 2H), 3.30 - 3.17 (m, 2H), 3.06 (br s, 4H), 2.44 - 2.09 (m, 4H), 2.09 - 1.51 (m,
5H); LC-MS: m/z 482.24 (MH+).
Preparation: (4-methyl-2-phenylpiperazin-l-yl)-[4-[(5-methyl-l,3,4-thiadiazol-2-yl)amino]-2- pyrrolidin-l-ylphenyl]methanone
Figure imgf000085_0001
Example 20
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 18 [4-[(3-methyl-l,2-oxazol-5-yl)amino]-2-pyrrolidin-l-ylphenyl]-(4-methyl-2- phenylpiperazin-l-yl)methanone wherein 5-methyl-l,3,4-thiadiazol-2-amine was used instead of 3- methyl-5-isoxazolamine. The title compound was obtained in a yield of 39 % (56 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.18 (d, 1H), 7.76 - 7.68 (m, 1H), 7.54 - 7.17 (m, 5H), 7.15 - 6.99
(m, 1H), 6.94 - 6.81 (m, 1H), 5.90 - 4.27 (m, 1H), 3.43 (d, 1H), 3.12 - 2.53 (m, 8H), 2.37 (dt, 1H), 2.22
(d, 4H), 2.07 - 1.84 (m, 4H), 1.81 - 1.55 (m, 2H); LC-MS: m/z 463.2 (MH+).
Preparation: (4-methyl-2-phenylpiperazin-l-yl)-[4-(lH-pyrazol-4-yl)-2-pyrrolidin-l- ylphenyl)methanone
Figure imgf000085_0002
Example 21 A mixture of (4-bromo-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone (50.0 mg, 0.120 mmol), lH-pyrazole-4-boronic acid (14.37 mg, 0.130 mmol) and a 2M aqueous solution of Na2CO3 (0.18 mL, 0.350 mmol) in 1,2-dimethoxyethane (1.052 mL) was degassed by 5 cycles of nitrogen/vacuum. Then palladium tetrakis triphenylphosphine (6.74 mg, 0.010 mmol) was added and the vessel was sealed. The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and then was heated to 95 °C for 2 days. After this time the mixture was directly purified by FC on
RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O
+ 0.1% of HCOOH) and then by SCX first washing with MeOH and then eluting with NH3 IN in MeOH.
Basic fractions were collected and concentrated under vacuum affording the product of formula (4- methyl-2-phenylpiperazin-l-yl)-[4-(lH-pyrazol-4-yl)-2-pyrrolidin-l-ylphenyl]methanone.
Yield: 14 mg
1H NMR (400 MHz, DMSO-d6) δ 12.91 (bs, 1H), 8.29 -8.15 (m, 1H), 7.98 -7.85 (m, 1H), 7.74 (d, 1H),
7.60 -7.15 (m, 4H), 7.08 -6.80 (m, 3H), 5.98 -4.37 (m, 1H), 3.52 -3.39 (m, 2H), 3.16 -2.61 (m, 5H), 2.44
-2.13 (m, 5H), 2.05 -1.90 (m, 3H), 1.83 -1.63 (m, 2H); LC-MS: m/z 416.18 (MH+).
Scheme 3
Figure imgf000086_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Enamine) by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
Alternatively compound of formula II may be obtained via acyl chloride formation of compound of formula I (commercially available from Enamine) by treatment with a suitable chlorinating agent, such as SOCl2, typically at 90 °C, followed by coupling with the proper amine in the presence of a suitable organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes from about 1 hour to about 12 hours to complete.
Step 2
Compound of formula III may be obtained by nitro reduction of compound of formula II using Zn dust in a suitable solvent, e.g. AcOH, typically at room temperature. The reaction takes from about
3 hours to about 12 hours to complete. Alternatively, the compound of formula III may be obtained by reduction of compound of formula II using Tin (III) chloride dehydrate in a suitable solvent, e.g.
EtOH, typically at 80 °C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula IV may be obtained by coupling reaction between compound of formula III and a proper commercially available acyl chloride, e.g. Cyclopropanecarbonyl chloride, in presence of an organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 3 hours to complete.
Step 4
Compound of formula V may be obtained via Suzuki coupling between compound of formula IV and the desired heteroaryl boronic ester or heteroaryl boronic acid, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2- dimethoxyethane / water, typically at 100 °C. The reaction takes about from 3 hours to 12 hours to complete.
Step 5
Compound of formula VI may be obtained via Miyaura coupling between compound of formula IV, the desired Aryl Bromide, and Bis(pinacolato)diboron, in presence of a suitable catalyst, such as PdCl2(dppf)*CH2Cl2, a suitable base, e.g. AcOK, in a suitable solvent, e.g. 1,4 dioxane, typically at 100
°C. The reaction takes about 12 hours to complete.
Step 6
Compound of formula V may be obtained via Suzuki coupling between compound of formula VI and commercially available heteroaryl halide in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na2CO3, in a suitable solvent, 1,2- dimethoxyethane, typically at 95 °C. The reaction takes about 12 hours to complete.
Scheme 3, Cpd (ll)*a Preparation: (2-bromo-4-nitrophenyl)-(4,4-difluoropiperidin-l-yl)methanone
Figure imgf000088_0001
To a solution of [dimethylamino(3-triazolo[4,5-b]pyridinyloxy)methylidene]-dimethylammonium hexafluorophosphate, HATU, (2.32 g, 6.1 mmol), 2-bromo-4-notrobenzoic acid (1.0 g, 4.06 mmol) and N,N-Diisopropylethylamine (3.15 g, 24.39 mmol) in DMF (25 mL), 4,4-difluoropiperidine hydrochloride (768.7 mg, 4.88 mmol) was added. The reaction was stirred at RT for 3h. After this time a saturated aqueous solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4, the solvent was filtered and evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from 100% of cHex to
AcOEt/cHex 25:75) affording the product of formula (2-bromo-4-nitrophenyl)-(4,4- difluoropiperidin-l-yl)methanone.
Yield: 1.2 g
1H NMR (400 MHz, DMSO) δ 8.51 (d, 1H), 8.32 (dd, 1H), 7.77 (d, 1H), 3.89 (dt, 1H), 3.68 (ddd, 1H),
3.25 (t, 2H), 2.32 - 1.88 (m, 4H); LC-MS: m/z 349.0, 351.0 (MH+)
Scheme 3, Cpd (ll)*b Preparation: (2-bromo-4-nitrophenyl)-(4-propylpiperazin-l-yl)methanone
Figure imgf000088_0002
A mixture of 2-bromo-4-nitrobenzoic acid (1.5 g, 6.1 mmol) and thionyl dichloride (8.9 mL, 121.95 mmol) was heated to 90 °C and stirred for 2 hours. The reaction was cooled down to RT and then concentrated under vacuum. The mixture was stripped three times with cyclohexane in order to remove thionyl dichloride excess. The residue was taken up with DCM (10 mL) and added to a solution of triethylamine (5.96 mL, 42.68 mmol) and 1-propylpiperazine dihydrobromide (1.77 g,
6.1 mmol) in DCM (75 mL).The reaction mixture was stirred for lh at RT, then it was diluted with
DCM, washed with a s.s. of NaHCO3, brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula (2-bromo-4-nitrophenyl)-(4-propylpiperazin-l- yl)methanone. The product was used in the next step without further purification.
Yield: 2.2 g
1H NMR (400 MHz, DMSO) δ 8.49 (d, 1H), 8.29 (dd, 1H), 7.65 (d, 1H), 3.65 (qdd, 2H), 3.18 - 3.05 (m,
2H), 2.49 - 2.38 (m, 2H), 2.37 - 2.31 (m, 2H), 2.30 - 2.24 (m, 2H), 1.44 (h, 2H), 0.86 (t, 3H); LC-MS: m/z 256.05, 358.07 (MH+).
Scheme 3, Cpd (ll)*c Preparation: (2-bromo-4-nitrophenyl)-(l,l-dioxo-l,4-thiazinan-4- yl)methanone
Figure imgf000089_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound (2- bromo-4-nitrophenyl)-(4-propylpiperazin-l-yl)methanone wherein 1,4-thiazinane 1,1-dioxide hydrochloride was used instead of 1-propylpiperazine dihydrobromide. The title compound was obtained in a yield of 74 % (3.3 g).
1H NMR (400 MHz, DMSO) δ 8.52 (d, 1H), 8.36 (dd, 1H), 7.89 (d, 1H), 4.50 - 4.36 (m, 1H), 3.74 (ddd,
1H), 3.60 - 3.51 (m, 2H), 3.42 (d, 1H), 3.28 - 3.23 (m, 1H), 3.12 - 3.05 (m, 2H); LC-MS: m/z 362.87,
364.94 (MH+).
Scheme 3, Cpd (ll)*d Preparation: (2-bromo-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000090_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound (2- bromo-4-nitrophenyl)-(4-propylpiperazin-l-yl)methanone wherein l-methyl-3-phenylpiperazine was used instead of 1-propylpiperazine dihydrobromide. The title compound was obtained in a yield of 100 % (1.8 g).
1H NMR (400 MHz, DMSO) δ 8.50 (td, 1H), 8.37 - 8.09 (m, 1H), 7.89 - 7.17 (m, 6H), 5.95 - 5.71 (m,
1H), 4.47 (d, 1H), 3.50 (dd, 1H), 3.17 - 2.57 (m, 2H), 2.40 (td, 1H), 2.30 - 1.90 (m, 4H); LC-MS: 404.01,
405.96 (MH+).
Scheme 3. Cpd (lll)*a Preparation: (4-amino-2-bromophenyl)-(4,4-difluoropiperidin-l- yl)methanone
Figure imgf000090_0001
To a solution of (2-bromo-4-nitrophenyl)-(4,4-difluoropiperidin-l-yl)methanone (1.17 g, 3.35 mmol) in AcOH (28 mL) zinc (2.19 g, 33.51 mmol) was added and the reaction mixture was stirred at RT for 24 h. The reaction was filtered and concentrated under vacuum. The residue was taken up with a s. s of NaHCO3 and extracted with AcOEt (3 times). The combined organic fractions were washed with brine, dried over Na2SO4, filtered and solvent was evaporated. The crude was purified by FC on silica gel (eluting from cHex/EtOAc 80:20 to cHex/EtOAc 4:6) affording the product of formula (4-amino-2-bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone.
Yield: 650 mg
NMR (400 MHz, DMSO-d6) δ 7.00 (d, 1H), 6.80 (d, 1H), 6.57 (dd, 1H), 5.62 (s, 2H), 3.86 - 3.73 (m,
2H), 3.67 - 3.55 (m, 2H), 2.13 - 1.91 (m, 4H); LC-MS: m/z 319.25, 321.25 (MH+).
Scheme 3. Cpd (lll)*b Preparation: (4-amino-2-bromophenyl)-(l,l-dioxo-l,4-thiazinan-4- yl)methanone
Figure imgf000091_0001
A mixture of (2-bromo-4-nitrophenyl)-(l,l-dioxo-l,4-thiazinan-4-yl)methanone (1.2 g, 3.3 mmol) and Tin(lll) chloride dihydrate (2.26 g, 9.91 mmol) in Ethanol (24 mL) was heated to reflux for 8 h. After this time the mixture was cooled down to RT, filtered and concentrated under vacuum.
The residue was purified by FC on silica gel (eluting from 100% of DCM to DCM/MeOH 95:5 as eluent) to obtain the product of formula (4-amino-2-bromophenyl)-(l,l-dioxo-l,4-thiazinan-4- yl)methanone.
Yield: 1.0 g
1H NMR (400 MHz, DMSO-d6) δ 7.10 (d, 1H), 6.80 (d, 1H), 6.57 (dd, 1H), 5.75 (s, 2H), 7H), 4.27 (s,
1H), 3.40 (d, 7H); LC-MS: m/z 332.93, 334.9 (MH+).
Scheme 3, Cpd (lll)*c Preparation: (4-amino-2-bromophenyl)-(4-propylpiperazin-l-yl)methanone
Figure imgf000091_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound (4- amino-2-bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone wherein (2-bromo-4-nitrophenylJ-
(4-propylpiperazin-l-yl)methanone was used instead of (2-bromo-4-nitrophenyl)-(4,4- difluoropiperidin-l-yl)methanone. The title compound was obtained in a yield of 88 % (1.7 g).
1H NMR (400 MHz, DMSO) δ 6.91 (d, 1H), 6.79 (d, 1H), 6.56 (dd, 1H), 5.57 (s, 2H), 3.57 (s, 2H), 3.15
(s, 2H), 2.35 (d, 3H), 2.29 - 2.14 (m, 3H), 1.43 (h, 2H), 0.85 (t, 3H); LC-MS: m/z 326.04, 327.99 (MH+).
Scheme 3, Cpd (lll)*d Preparation: (4-amino-2-bromophenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000091_0003
The synthesis of the title compound was effected analogously to the synthesis of the compound (4- amino-2-bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone wherein (2-bromo-4-nitrophenyl)-
(4-methyl-2-phenylpiperazin-l-yl)methanone was used instead of (2-bromo-4-nitrophenyl)-(4,4- difluoropiperidin-l-yl)methanone. The title compound was obtained in a yield 91 % (850 mg).
1H NMR (400 MHz, DMSO) δ 7.57 (d, 1H), 7.30 (d, 5H), 6.80 (td, 2H), 5.75 (s, 1H), 5.60 (s, 2H), 4.95
- 4.27 (m, 1H), 3.45 (d, 1H), 3.23 - 2.87 (m, 2H), 2.77 - 2.57 (m, 1H), 2.38 - 2.13 (m, 3H), 1.89 (s, 1H);
LC-MS: m/z 374.09, 376.08 (MH+).
Scheme 3, Cpd (IV)*a Preparation: N-[3-bromo-4-(4,4-difluoropiperidine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000092_0001
To a cooled mixture of N,N-Diisopropylethylamine (0.69 mL, 4.09 mmol) and (4-amino-2- bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone (652.0 mg, 2.04 mmol) in DCM (20 mL), at 0
°C, cyclopropanecarbonyl chloride (0.22 mL, 2.45 mmol) was added dropwise. The mixture was stirred for lh at RT. After this time, the mixture was diluted with DCM, washed with a s.s. of NaHCO3 and dried over a phase separator. The solvent was evaporated and crude was purified by FC on silica gel (eluting from 100% of DCM to DCM/MeOH 95:5) affording the product of formula N-[3-bromo-
4-(4,4-difluoropiperidine-l-carbonyl)phenyl]cyclopropanecarboxamide.
Yield: 830 mg
1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 8.03 (d, 1H), 7.56 (dd, 1H), 7.33 (d, 1H), 3.82 (s, 1H), 3.66
(d, 1H), 3.24 (t, 2H), 2.17 - 1.85 (m, 4H), 1.82 - 1.71 (m, 1H), 0.87 - 0.80 (m, 4H); LC-MS: m/z 387.16,
389.16 (MH+).
Scheme 3, Cpd (IV)*b Preparation: N-[3-bromo-4-(4-propylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000092_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)phenyl]cyclopropanecarboxamide wherein (4- amino-2-bromophenyl)-(4-propylpiperazin-l-yl)methanone was used instead of (4-amino-2- bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone. The title compound was obtained in a yield
96 % (2.0 g).
1H NMR (400 MHz, DMSO) δ 10.45 (s, 1H), 8.02 (d, 1H), 7.55 (ddlH), 7.24 (d, 1H), 3.61 (d, 2H), 3.12
(td, 2H), 2.46 - 2.09 (m, 6H), 1.77 (p, 1H), 1.42 (dt, 2H), 0.92 - 0.78 (m, 7H); LC-MS: m/z 394.10,
396.04 (MH+).
Scheme 3. Cpd (IV)*c Preparation: N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000093_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)phenyl]cyclopropanecarboxamide wherein (4- amino-2-bromophenyl)-(l,l-dioxo-l,4-thiazinan-4-yl)methanone was used instead of (4-amino-2- bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone. The title compound was obtained in a yield
43 % (1.6 g).
1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H), 8.04 (d, 1H), 7.59 (dd, 1H), 7.46 (d, 1H), 4.35 (s, 1H), 3.75
(s, 1H), 3.56 (s, 3H), 3.23 (s, 2H), 3.06 (s, 1H), 1.78 (p, 1H), 0.84 (d, 4H); LC-MS: m/z 401.27, 403.27
(MH+).
Scheme 3. Cpd (IV)*d Preparation: N-[3-bromo-4-(4-methyl-2-phenylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000093_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)phenyl]cyclopropanecarboxamide wherein (4- amino-2-bromophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone was used instead of (4- amino-2-bromophenyl)-(4,4-difluoropiperidin-l-yl)methanone. The title compound was obtained in a yield of 69 % (3.9 g).
NMR (400 MHz, DMSO-d6) δ 10.44 (d, 1H), 8.14 - 7.94 (m, 1H), 7.58 (d, 2H), 7.31 (dt, 5H), 5.84 -
5.52 (m, 1H), 3.47 (d, 1H), 3.17 - 2.57 (m, 2H), 2.46 - 2.12 (m, 4H), 2.11 - 1.60 (m, 2H), 1.08 (dt, 1H),
0.82 (d, 4H); LC-MS: m/z 442.12, 444.14 (MH+).
Preparation: N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)phenyl]cyclopropanecarboxamide
Figure imgf000094_0001
Example 22
A mixture of l-methyl-lH-pyrazole-3-boronic acid pinacol ester (83.83 mg, 0.400 mmol), N-[3- bromo-4-(4,4-difluoropiperidine-l-carbonyl)phenyl]cyclopropanecarboxamide (120.0 mg, 0.310 mmol), X-Phos amonobiphenyl palladium chloride precatalyst (24.35 mg, 0.030 mmol) and tripotassium phosphate (197.35 mg, 0.930 mmol) in Water (0.3 mL) / 1,2-dimethoxyethane (3 mL was degassed under nitrogen flux for 5 min. and then heated to 100 °C overnight. The day after the reaction was cooled down to room temperature, diluted with water and extracted with EtOAc (3x).
The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The residue was purified by FC on silica gel (eluting from
DCM/EtOAc 90:10 to DCM:AcOEt 20:80) and again by FC on reverse phase using acid conditions
(eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of
NaHCO3 and extracted 3 times with AcOEt. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula N-[4-
(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3-yl)phenyl]cyclopropanecarboxamide.
Yield: 22 mg
1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H), 7.95 (d, 1H), 7.73 (d, 1H), 7.63 (dd, 1H), 7.20 (d, 1H), 6.35
(d, 1H), 3.85 (s, 4H), 3.51 (ddd, 1H), 3.13 (tt, 2H), 2.13 - 1.89 (m, 2H), 1.79 (tt, 2H), 1.60 - 1.47 (m,
1H), 0.85 - 0.75 (m, 4H); LC-MS: m/z 389.2 (MH+) Preparation: N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide
Figure imgf000095_0001
Example 23
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 22 N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein 1-isopropyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole was used instead of l-methyl-lH-pyrazole-3-boronic acid pinacol ester. The title compound was obtained in a yield of 42 % (54 mg).
1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H), 7.91 (d, 1H), 7.80 (d, 1H), 7.65 (dd, 1H), 7.21 (d, 1H), 6.34
(d, 1H), 4.51 (hept, 1H), 3.93 - 3.82 (m, 1H), 3.51 (ddd, 1H), 3.20 - 3.02 (m, 2H), 2.14 - 1.66 (m, 4H),
1.42 (dd, 6H), 1.36 - 1.21 (m, 1H), 0.85 - 0.75 (m, 4H); LC-MS: m/z 417.3 (MH+).
Preparation: N-[3-(l-propan-2-ylpyrazol-3-yl)-4-(4-propylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000095_0002
Example 24
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 22 N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein N-[3-bromo-4-(4-propylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide and l-isopropyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole were used instead of N-[3-bromo-4-(4,4-difluoropiperidine-l- carbonyl)phenyl]cyclopropanecarboxamide and l-methyl-lH-pyrazole-3-boronic acid pinacol ester respectively. The title compound was obtained in a yield of 38 % (61 mg). 1H NMR (400 MHz, DMSO) δ 10.34 (s, 1H), 7.91 (d, 1H), 7.78 (d, 1H), 7.64 (dd, 1H), 7.13 (d, 1H), 6.30
(d, 1H), 4.52 (hept, 1H), 3.62 - 3.43 (m, 2H), 3.06 - 2.96 (m, 1H), 2.90 - 2.78 (m, 1H), 2.30 (dt, 2H),
2.20 - 2.03 (m, 3H), 1.88 - 1.66 (m, 2H), 1.44 (dd, 6H), 1.36 (h, 2H), 0.84 - 0.74 (m, 7H); LC-MS: m/z
424.3 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide
Figure imgf000096_0001
Example 25
4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2-dioxaborolane (72.61 mg, 0.320 mmol), N-[3- bromo-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide (100.0 mg, 0.250 mmol), X-Phos amonobiphenyl palladium chloride precatalyst (19.58 mg, 0.020 mmol) and tripotassium phosphate (158.69 mg, 0.750 mmol) were suspended in a mixture of Water (0.3 mL) and 1,2-dimethoxyethane (3.3 mL). The resulting suspension was degassed under nitrogen flux for
5 min and then heated to 90 °C overnight. The day after the reaction mixture was cooled down to room temperature, diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from
5:95 of CH3CN/H2O + 0.1% of HCOOH to 60:40 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide.
Yield: 23 mg
1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 7.83 (d, 1H), 7.59 (dd, 1H), 7.35 (d, 1H), 6.94 (d, 1H), 6.84
(dt, 1H), 4.06 - 3.98 (m, 1H), 3.97 - 3.90 (m, 1H), 3.53 - 3.44 (m, 1H), 3.33 (d, 1H), 3.10 - 2.97 (m, 2H),
2.50 (d, 1H), 2.47 (d, 3H), 2.28 (d, 1H), 1.84 - 1.73 (m, 1H), 0.85 - 0.78 (m, 4H); LC-MS: m/z 419.1
(MH+).
Preparation: N-[3-(5-cyanothiophen-2-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000097_0001
Example 26
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein (5-cyanothiophen-2-yl)boronic acid was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2-dioxaborolane. The title compound was obtained in a yield of 11 % (11 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 7.95 (dd, 2H), 7.72 (dd, 1H), 7.53 (d, 1H), 7.26 (d, 1H),
4.27 (s, 1H), 3.55 (dd, 3H), 3.12 (s, 3H), 2.67 (p, 1H), 1.79 (p, 1H), 0.84 (d, 4H); LC-MS: m/z 430.2
(MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-[2-(trifluoromethyl)-l,3-thiazol-5- yl]phenyl]cyclopropanecarboxamide
Figure imgf000097_0002
Example 27
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein 5-(tetramethyl-l,3,2-dioxaborolan-2-yl)-2-
(trifluoromethyl)-l,3-thiazole was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-
1,3,2-dioxaborolane. The title compound was obtained in a yield of 55 % (62 mg).
1H NMR (400 MHz, DMSO-d6) δ10.56 (s, 1H), 8.13 (q, 1H), 7.97 (d, 1H), 7.74 (dd, 1H), 7.61 (d, , 1H),
4.37 (s, 1H), 3.57 (s, 3H), 3.30 (s, 4H), 1.80 (p, 1H), 0.88 - 0.81 (m, 4H); LC-MS: m/z 474.1 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide
Figure imgf000098_0002
Example 28
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-
1,3,2-dioxaborolane. The title compound was obtained in a yield of 37 % (38 mg).
NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 7.91 (d, 1H), 7.84 (d, 1H), 7.66 (dd, 1H), 7.28 (d, 1H),
6.38 (d, 1H), 4.52 (p, 1H), 4.09 (s, 1H), 3.97 - 3.86 (m, 1H), 3.56 - 3.36 (m, 2H), 3.10 - 3.00 (m, 2H),
2.58 - 2.53 (m, 2H), 1.80 (p, 1H), 1.44 (dd, 6H), 0.85 - 0.68 (m, 4H); LC-MS: m/z 431.2 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-[5-(trifluoromethyl)thiophen-2- yl)phenyl)cyclopropanecarboxamide
Figure imgf000098_0001
Example 29
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 25 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide wherein [5-(trifluoromethyl)thiophen-2-yl]boronic acid was used instead of 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2-dioxaborolane. The title compound was obtained in a yield of 11 % (12 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.54 (s, 1H), 7.96 (d, 1H), 7.76 - 7.68 (m, 2H), 7.51 (d, 1H), 7.22
(dq, 1H), 4.21 (s, 1H), 3.74 (s, 1H), 3.56 (s, 1H), 3.32 (s, 3H), 3.17 (d, 1H), 3.04 (s, 1H), 1.79 (p, 1H),
0.87 - 0.80 (m, 4H); LC-MS: m/z 473.1 (MH+). Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide
Figure imgf000099_0001
Example 30
A mixture of X-Phos aminobiphenyl palladium chloride precatalyst (10.94 mg, 0.010 mmol), tripotassium phosphate (88.68 mg, 0.420 mmol), l-propan-2-yl-3-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyrazole (32.88 mg, 0.140 mmol) and N-[3-bromo-4-(4-methyl-2- phenylpiperazine-l-carbonyl)phenyl]cyclopropanecarboxamide (80.0 mg, 0.140 mmol) in Water
(0.195 mL)/l,2-dimethoxyethane (1.945 mL) was degassed for 5 min and then stirred at 100 °C ON.
The day after the mixture was diluted with water and extracted with EtOAc x3. The combined organic phases were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on reverse phase using basic conditions (eluting from 5:95 of CH3CN/H2O + 0.1 % of NH4OH to 40:60 of CH3CN/H2O + 0.1 % of NH4OH) and again by FC on NH column (eluting from 100% of DCM to DCM/EtOAc 60:40) affording the product of formula N-[4-(4- methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide as a racemic mixture.
Yield: 24 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 10.42 - 10.28 (m, 1H), 8.00 - 6.93 (m, 10H), 6.47 - 5.66 (m, 3H),
4.70 - 4.30 (m, 3H), 3.41 - 3.33 (m, 1H), 3.10 - 2.55 (m, 4H), 2.41 - 1.72 (m, 2H), 1.56 - 1.00 (m, 5H),
0.86 - 0.75 (m, 4H); LC-MS: m/z 472.28 (MH+).
Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-[2-(trifluoromethyl)-l,3-thiazol-5- yl]phenyl]cyclopropanecarboxamide
Figure imgf000100_0001
Example 31
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 30 N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein 5-(tetramethyl-l,3,2-dioxaborolan-2-yl)-2-
(trifluoromethyl)-l,3-thiazole was used instead of l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole. The title compound was obtained in a yield of 14 % (10 mg) as a racemic mixture.
1H NMR (400 MHz, DMSO-d6) δ ppm 10.64 - 10.49 (m, 1H), 8.38 - 7.12 (m, 8H), 5.80 - 5.64 (m, 1H),
4.43 - 4.32 (m, 1H), 3.35 - 3.28 (m, 2H), 2.44 - 1.70 (m, 8H), 0.88 - 0.72 (m, 4H); LC-MS: m/z 515.17
(MH+).
Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(2-methyl-l,3-thiazol-5- yl)phenyl]cyclopropanecarboxamide
Figure imgf000100_0002
Example 32
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 30 N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein 2-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-
2-yl)-l,3-thiazole was used instead of l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazole. The title compound was obtained in a yield of 47 % (19 mg) as a racemic mixture.
NMR (400 MHz, DMSO-d6) δ 10.54 - 10.34 (m, 1H), 7.94 - 7.60 (m, 2H), 7.55 - 7.16 (m, 6H), 5.73
- 5.64 (m, 1H), 4.60 - 4.28 (m, 2H), 3.12 - 2.53 (m, 5H), 2.02 (s, 6H), 1.17 - 0.90 (m, 1H), 0.89 - 0.74
(m, 4H); LC-MS: m/z 461.25 (MH+). Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-[5-(trifluoromethyl)thiophen-2- yl]phenyl]cyclopropanecarboxamide
Figure imgf000101_0001
Example 33
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 30 N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein [5-(trifluoromethyl)thiophen-2-yl]boronic acid was used instead of l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole. The title compound was obtained in a yield of 20 % (11 mg) as a racemic mixture.
1H NMR (400 MHz, DMSO-d6) δ ppm 10.56 - 10.41 (m, 1H), 8.04 - 7.15 (m, 10H), 5.78 - 5.66 (m, 1H),
4.55 - 4.28 (m, 1H), 3.39 - 3.25 (m, 1H), 3.13 - 2.54 (m, 3H), 2.41 - 2.27 (m, 1H), 2.23 - 2.12 (m, 2H),
2.05 - 1.73 (m, 1H), 1.57 - 1.45 (m, 1H), 0.83 (br d, 4H); LC-MS: m/z 514.29 (MH+).
Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(5-methylthiophen-2- yl)phenyl]cyclopropanecarboxamide
Figure imgf000101_0002
Example 34
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 3.1 N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)phenyl]cyclopropanecarboxamide wherein 4,4,5,5-tetramethyl-2-(5-methyl-2-thiophenyl)-l,3,2- dioxaborolane was used instead of l-propan-2-yl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazole. The title compound was obtained in a yield of 61 % (38 mg) as a racemic mixture. 1H NMR (400 MHz, DMSO-d6) δ 10.48 - 10.26 (m, 1H), 7.90 - 7.56 (m, 2H), 7.50 - 7.33 (m, 2H), 7.25
(dtd, 4H), 7.09 - 6.55 (m, 2H), 5.86 - 4.17 (m, 1H), 3.38 - 3.29 (m, 2H), 3.08 - 2.56 (m, 3H), 2.45 - 2.11
(m, 3H), 2.05 - 1.52 (m, 4H), 1.38 - 0.74 (m, 5H); LC-MS: m/z 460.18 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl]cyclopropanecarboxamide
Figure imgf000102_0001
Example 35
A mixture of N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide
(100.0 mg, 0.240 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,3,2- dioxaborolane (74.42 mg, 0.290 mmol), potassium acetate (72.64 mg, 0.730 mmol) and [1,1'-
Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (4.0 mg, 0 mmol) in 1,4-Dioxane (2.442 mL) was degassed using Shlenk line technique and stirred at 100
°C overnight. The day after the reaction was cooled down to RT and filtered through a pad of celite washing with EtOAc. The solution was concentrated under vacuum affording the product of formula
N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]cyclopropanecarboxamide, which was used in the next step without further purification.
Yield: 100 mg
1H NMR (400 MHz, DMSO) δ 10.37 (d, 1H), 7.81 (dd, 1H), 7.70 -7.60 (m, 1H), 7.44 (d, 1H), 3.57 (s,
8H), 1.84 -1.77 (m, 1H), 1.27 (s, 6H), 1.07 (s, 6H), 0.90 -0.76 (m, 4H); LC-MS: m/z 449.21 (MH+).
Preparation: N-[4-(2-oxa-7-azaspiro[3.5]nonane-7-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl]cyclopropanecarboxamide
Figure imgf000102_0002
Example 36 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 3. N N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-
2-yl)phenyl]cyclopropanecarboxamide wherein N-[3-bromo-4-(2-oxa-7-azaspiro[3.5]nonane-7- carbonyl)phenyl]cyclopropanecarboxamide was used instead of N-[3-bromo-4-(l,l-dioxo-l,4- thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide. The title compound was obtained in a quantitative yield (80 mg).
LC-MS: m/z 441.25 (MH+)
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-[6-(trifluoromethyl)pyridin-2- yl]phenyl]cyclopropanecarboxamide
Figure imgf000103_0001
Example 37
Palladium tetrakis triphenylphosphine (14.43 mg, 0.010 mmol) and N-[4-(l,l-dioxo-l,4-thiazinane-
4-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]cyclopropanecarboxamide
(112.0 mg, 0.250 mmol) were suspended in 1,2-dimethoxyethane (2.5 mL) and then a 2 M aqueous solution of sodium carbonate (0.37 mL, 0.750 mmol) was added. The mixture was degassed by 5 cycles of nitrogen/vacuum and 2-bromo-6-(trifluoromethyl)pyridine (62.1 mg, 0.270 mmol) was added. The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and heated to 90
°C overnight. The day after the reaction mixture was cooled down to room temperature and diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The mixture was purified by FC on RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to
35:65 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula N-[4-(l,l-dioxo-l,4- thiazinane-4-carbonyl)-3-[6-(trifluoromethyl)pyridin-2-yl]phenyl]cyclopropanecarboxamide.
Yield: 22 mg
1H NMR (400 MHz, DMSO) δ 10.52 (s, 1H), 8.21 (t, 1H), 8.04 (d, 1H), 7.95 (d, 1H), 7.89 (dd, 1H), 7.75
(dd, 1H), 7.55 (d, 1H), 4.37 (s, 1H), 3.30 (s, 7H), 1.81 (p, 1H), 0.84 (d, 4H); LC-MS: m/z 468.17 (MH+). Preparation: N-[4-(2-oxa-7-azaspiro[3.5]nonane-7-carbonyl)-3-[6-(trifluoromethyl)pyridin-2- yl]phenyl]cyclopropanecarboxamide
Figure imgf000104_0001
Example 38
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 37 N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-[6-(trifluoromethyl)pyridin-2- yl]phenyl]cyclopropanecarboxamide wherein N-[4-(2-oxa-7-azaspiro[3.5]nonane-7-carbonyl)-3-
(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]cyclopropanecarboxamide was used instead of N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl]cyclopropanecarboxamide. The title compound was obtained in a yield of 23 % (20 mg).
1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H), 8.17 (t, 1H), 7.98 (d, 1H), 7.86 (dd, 2H), 7.74 (dd, 1H),
7.30 (d, 1H), 4.44 -4.07 (m, 4H), 3.30 (s, 4H), 1.81 (p, 1H), 1.76 -1.37 (m, 4H), 0.88 -0.79 (m, 4H); LC-
MS: m/z 460.20 (MH+).
Scheme 4
Figure imgf000104_0002
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Astatech) by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete. Step 2
Compound of formula III may be obtained via Suzuki coupling between compound of formula II and the desired heteroaryl boronic ester, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 100 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Step 3
Compound of formula IV may be obtained by nitrile cyclization of compound of formula III with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Step 4
Compound of formula V may be obtained by nitrile cyclization of compound of formula III with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Step 5
Compound of formula IV may be obtained via Suzuki coupling between compound of formula V and the desired heteroaryl boronic ester, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 100 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Scheme 4, Cpd (ll)*a Preparation: 3-bromo-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile
Figure imgf000105_0001
To a solution of HATU, (0.63 g, 1.66 mmol), 2-bromo-4-cyanobenzoic acid (250.0 mg, 1.11 mmol) and N,N-Diisopropylethylamine (1.13 mL, 6.64 mmolJ in DMF (7.374 mL), 4-methoxy-4-methyl- piperidine hydrochloride (219.87 mg, 1.33 mmol) was added. The reaction was stirred at RT for 2.5 h. After this time a saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from cHex/AcOEt 95:5 to cHex/AcOEt 50:50) affording the product of formula 3-bromo-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile.
Yield: 170 mg
1H NMR (400 MHz, DMSO) δ 8.27 (t, 1H), 7.94 (ddd, 1H), 7.62 - 7.50 (m, 1H), 4.13 (ddd, 1H), 3.22 -
3.02 (m, 5H), 3.00 - 2.89 (m, 1H), 1.86 - 1.72 (m, 1H), 1.68 - 1.57 (m, 1H), 1.57 - 1.33 (m, 2H), 1.12
(d, 3H); LC-MS: m/z 337.25, 339.25 (MH+).
Scheme 4, Cpd (ll)*b Preparation: 3-bromo-4-(4-tert-butylpiperidine-l-carbonyl)benzonitrile
Figure imgf000106_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-tert-butylpiperidine was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 90 % (346 mg).
1H NMR (400 MHz, DMSO-d6) δ 8.30 - 8.25 (m, 1H), 7.98 - 7.87 (m, 1H), 7.66 - 7.47 (m, 1H), 4.59 (t,
1H), 3.23 - 3.11 (m, 1H), 3.02 - 2.91 (m, 1H), 2.76 - 2.63 (m, 1H), 1.90 - 1.50 (m, 2H), 1.34 - 1.03 (m,
3H), 0.89 - 0.81 (m, 9H); LC-MS: m/z 349.17, 351.13 (MH+).
Scheme 4, Cpd (ll)*c Preparation: 3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile
Figure imgf000106_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4,4-difluoropiperidine hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 77 % (500 mg).
NMR (400 MHz, DMSO-d6) δ 8.31 (d, 1H), 7.98 (dd, 1H), 7.66 (d, 1H), 3.95 - 3.83 (m, 1H), 3.78 -
3.61 (m, 1H), 3.23 (t2H), 2.23 - 1.87 (m, 4H); LC-MS: m/z 329.25, 331.25 (MH+). Scheme 4, Cpd (ll)*d Preparation: 3-bromo-4-(l-oxa-7-azaspiro[3.5]nonane-7- carbonyl)benzonitrile
Figure imgf000107_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein l-oxa-7- azaspiro[3.5]nonane was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 74 % (487 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.27 (d, 1H), 7.93 (dt, 1H), 7.56 (dd, 1H), 4.46 - 4.33 (m, 2H),
3.73 - 3.51 (m, 2H), 3.27 (s, 1H), 2.99 (s, 2H), 2.37 (br d, 2H), 1.96 - 1.70 (m, 3H); LC-MS: m/z 335.09,
337.06 (MH+).
Scheme 4, Cpd (ll)*e Preparation: 3-bromo-4-(4-ethoxypiperidine-l-carbonyl)benzonitrile
Figure imgf000107_0003
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-ethoxipiperidine hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 56 % (210 mg).
1H NMR (400 MHz, DMSO) δ 8.28 (t, 1H), 7.94 (dd, 1H), 7.56 (dd, 1H), 3.97 (td, 1H), 3.54 (tt, 1H),
3.46 (qd, 2H), 3.35 - 3.31 (m, 1H), 3.19 (ddt, 1H), 2.99 (tdd, 1H), 1.89 (s, 1H), 1.81 - 1.72 (m, 1H),
1.58 - 1.31 (m, 2H), 1.10 (t, 3H); LC-MS: m/z 337.35, 339.35 (MH+).
Scheme 4, Cpd Preparation: 3-bromo-4-(6,6-difluoro-2-azaspiro[3.3]heptane-2- carbonyl)benzonitrile
Figure imgf000107_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 6,6-difluoro-2- azaspiro[3.3]heptane hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 50 % (70 mg).
NMR (400 MHz, DMSO-d6) δ 8.30 (d, 1H), 7.96 (dd, 1H), 7.61 (d, 1H), 4.19 (s, 2H), 3.97 (s, 2H),
2.96 - 2.78 (m, 4H); LC-MS: m/z 341.25, 343.25 (MH+)
Scheme 4. Cpd (ll)*g Preparation: 3-bromo-4-[4-(trifluoromethoxy)piperidine-l- carbonyl)benzonitrile
Figure imgf000108_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-
(trifluoromethoxy)piperidine hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 62 % (260 mg).
1H NMR (400 MHz, DMSO) δ 8.29 (dd, 1H), 7.96 (ddd, 1H), 7.72 - 7.49 (m, 1H), 4.74 (dt, 1H), 4.02
(td, 1H), 3.42 (dddd, 1H), 3.30 - 3.06 (m, 2H), 2.09 - 1.97 (m, 1H), 1.90 (s, 1H), 1.84 - 1.56 (m, 2H);
LC-MS: m/z 377.26, 379.26 (MH+).
Scheme 4, Cpd (ll)*h Preparation: 3-bromo-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile
Figure imgf000108_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-
(trifluoromethoxy)piperidine hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 78 % (560 mg). 1H NMR (400 MHz, DMSO) δ 8.29 (dd, 1H), 7.95 (ddd, 1H), 7.71 - 7.50 (m, 1H), 4.60 (t, 1H), 3.26 (d,
1H), 3.09 (dtd, 1H), 2.83 (tddlH), 2.66 (d, 1H), 1.94 (t, 1H), 1.82 - 1.65 (m, 1H), 1.55 - 1.29 (m, 2H);
LC-MS: m/z 361.10, 363.09 (MH+).
Scheme 4, Cpd (ll)*i Preparation: 3-bromo-4-(4-fluoro-4-methylpiperidine-l-carbonyl)benzonitrile
Figure imgf000109_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-fluoro-4- methylpiperidine hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride.
The title compound was obtained in a yield of 52 % (186 mg).
1H NMR (400 MHz, DMSO) δ 8.29 (d, 1H), 7.95 (ddd, 1H), 7.65 - 7.53 (m, 1H), 4.26 (d, 1H), 3.25 -
2.97 (m, 3H), 1.94 - 1.79 (m, 1H), 1.78 - 1.54 (m, 3H), 1.42 - 1.28 (m, 3H); LC-MS: m/z 325.25, 327.25
(MH+).
Scheme 4, Cpd (ll)*i Preparation: 3-bromo-4-(4-methoxypiperidine-l-carbonyl)benzonitrile
Figure imgf000109_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-methoxypiperidine was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 56 % (200 mg).
1H NMR (400 MHz, DMSO) δ 8.28 (d, 1H), 7.94 (dt, 1H), 7.56 (dd, 1H), 3.97 - 3.87 (m, 1H), 3.49 - 3.33
(m, 2H), 3.25 (s, 3H), 3.18 (qd, 1H), 3.06 - 2.92 (m, 1H), 1.95 - 1.84 (m, 1H), 1.76 (s, 1H), 1.59 - 1.34
(m, 2H); LC-MS: 323.25, 325.25 (MH+).
Scheme 4, Cpd (ll)*k Preparation 3-bromo-4-(4-methylpiperidine-l-carbonyl)benzonitrile
Figure imgf000109_0003
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-methylpiperidine was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 78 % (265 mg).
1H NMR (400 MHz, DMSO) δ 8.27 (dd, 1H), 7.93 (ddd, 1H), 7.60 - 7.46 (m, 1H), 4.45 (t, 1H), 3.14 (d,
1H), 3.07 - 2.91 (m, 1H), 2.78 (tt, 1H), 1.76 - 1.45 (m, 3H), 1.26 - 0.97 (m, 2H), 0.91 (d, 3H); LC-MS: m/z 307.10, 309.09 (MH+).
Scheme 4, Cpd (I l)*l Preparation: 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-bromobenzonitrile
Figure imgf000110_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 6-azaspiro[2.5]octane hydrochloride was used instead of 4-methoxy-4-methyl-piperidine hydrochloride. The title compound was obtained in a yield of 78 % (550 mg).
NMR (400 MHz, DMSO-d6) δ 8.28 (d, 1H), 7.94 (dd, 1H), 7.57 (d, 1H), 3.67 (t, 2H), 3.16 - 3.05 (m,
2H), 1.56 - 1.20 (m, 4H), 0.51 - 0.24 (m, 4H); LC-MS: m/z 319.14, 321.10 (MH+).
Scheme 4, Cpd (lll)*a Preparation: 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine- l-carbonyl)benzonitrile
Figure imgf000110_0001
A mixture of l-(propan-2-yl)-3-(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (138.64 mg,
0.590 mmol), 3-bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile (180.0 mg, 0.530 mmol), X-phos amonobiphenyl palladium chloride precatalyst (41.95 mg, 0.050 mmol), tripotassium phosphate (339.92 mg, 1.6 mmol) in Water (0.755 mL) / 1,2-dimethoxyethane (7.554 mL) was degassed under nitrogen flux for 5 min and then heated to 90 °C for lh. After this time the reaction mixture was cooled down to room temperature, diluted with water and extracted with AcOEt (3x).
The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The residue was purified by FC on silica gel (eluting form 100
% of cHex to AcOEt/cHex 30:70) affording the product of formula 3-(l-isopropylpyrazol-3-yl)-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile.
Yield: 120 mg
1H NMR (400 MHz, DMSO) δ 8.23 - 8.05 (m, 1H), 7.89 - 7.76 (m, 2H), 7.44 (dd, 1H), 6.69 - 6.50 (m,
1H), 4.52 (dp, 1H), 4.19 - 4.08 (m, 1H), 3.09 - 2.79 (m, 6H), 1.75 (t, 1H), 1.52 - 1.26 (m, 9H), 1.10 -
0.95 (m, 3H); LC-MS: m/z 367.5 (MH+).
Scheme 4, Cpd (lll)*b Preparation: 4-(4-tert-butylpiperidine-l-carbonyl)-3-(l-isopropylpyrazol-3- yl)benzonitrile
Figure imgf000111_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4-tert-butylpiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 91 % (100 mg).
NMR (400 MHz, DMSO-d6) δ 8.24 - 8.06 (m, 1H), 7.87 - 7.76 (m, 2H), 7.49 - 7.37 (m, 1H), 6.64 -
6.47 (m, 1H), 4.71 - 4.40 (m, 2H), 3.28 - 3.01 (m, 2H), 2.91 - 2.53 (m, 2H), 1.83 - 1.63 (m, 1H), 1.57 -
1.38 (m, 7H), 1.32 - 1.13 (m, 2H), 0.91 - 0.60 (m, 9H); LC-MS: m/z 379.36 (MH+).
Scheme 4, Cpd (lll)*c Preparation: 3-(l-isopropylpyrazol-3-yl)-4-(l-oxa-7-azaspiro[3.5]nonane-7- carbonyl)benzonitrile
Figure imgf000111_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(l-oxa-7-azaspiro[3.5]nonane-7-carbonyl)benzonitrile was used instead of 3-bromo-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 60 % (65 mg).
NMR (400 MHz, DMSO-d6) δ 8.16 (dd, 1H), 7.86 - 7.80 (m, 2H), 7.45 (dd, 1H), 6.59 (dd, 1H), 4.57
- 4.28 (m, 2H), 3.71 - 3.50 (m, 2H), 3.15 - 2.79 (m, 2H), 2.43 - 2.18 (m, 2H), 1.90 - 1.73 (m, 2H), 1.63
- 1.55 (m, 1H), 1.49 - 1.36 (m, 7H), 1.32 - 1.21 (m, 1H); LC-MS: m/z 365.24 (MH+).
Scheme 4, Cpd (lll)*d Preparation: 4-(4-ethoxypiperidine-l-carbonyl)-3-(l-isopropylpyrazol-3- yl)benzonitrile
Figure imgf000112_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4-ethoxypiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-(4-methoxy-
4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 79 %
(180 mg).
1H NMR (400 MHz, DMSO) δ 8.16 (dd, 1H), 7.88 - 7.77 (m, 2H), 7.44 (t, 1H), 6.58 (t, 1H), 4.53 (dp,
1H), 4.00 - 3.84 (m, 1H), 3.44 (qd, 2H), 3.40 - 2.99 (m, 3H), 2.94 - 2.73 (m, 1H), 1.88 - 1.75 (m, 1H),
1.59 - 1.49 (m, 2H), 1.43 (td, 6H), 1.34 - 1.18 (m, 1H), 1.07 (dt, 3H); LC-MS: m/z 367.3 (MH+).
Scheme 4, Cpd (lll)*e Preparation: 4-(6,6-difluoro-2-azaspiro[3.3]heptane-2-carbonyl)-3-(l- isopropylpyrazol-3-yl)benzonitrile
Figure imgf000112_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(6,6-difluoro-2-azaspiro[3.3]heptane-2-carbonyl)benzonitrile was used instead of 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 60 % (52 mg).
LC-MS: m/z 371.46 (MH+)
Scheme 4, Cpd (lll)*f Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)benzonitrile
Figure imgf000113_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 100 % (90 mg).
NMR (400 MHz, DMSO-d6) δ 8.22 - 8.17 (m, 1H), 7.89 - 7.82 (m, 2H), 7.57 - 7.47 (m, 1H), 6.65 (d,
1H), 4.51 (p, 1H), 3.80 - 3.66 (m, 2H), 3.25 - 2.99 (m, 2H), 2.16 - 1.77 (m, 2H), 1.54 (dq, 1H), 1.43 (dd,
7H); LC-MS: m/z 359.2 (MH+).
Scheme 4, Cpd (lll)*g reparation: 4-(4-fluoro-4-methylpiperidine-l-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile
Figure imgf000113_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4-fluoro-4-methylpiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 100 % (204 mg).
1H NMR (400 MHz, DMSO) δ 8.22 - 8.08 (m, 1H), 7.89 - 7.78 (m, 2H), 7.47 (dd, 1H), 6.67 - 6.51 (m,
1H), 4.52 (h, 1H), 4.33 - 4.17 (m, 1H), 3.13 - 2.88 (m, 3H), 1.84 - 1.12 (m, 13H); LC-MS: m/z 355.2
(MH+).
Scheme 4, Cpd (lll)*h Preparation: 3-(l-propan-2-ylpyrazol-3-yl)-4-[4-(trifluoromethyl)piperidine- l-carbonyl]benzonitrile
Figure imgf000114_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-bromo-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 100 % (220 mg).
1H NMR (400 MHz, DMSO) δ 8.25 - 8.06 (m, 1H), 7.88 - 7.77 (m, 2H), 7.56 - 7.40 (m, 1H), 6.67 - 6.47
(m, 1H), 4.62 (s, 1H), 4.57 - 4.46 (m, 1H), 3.26 - 2.62 (m, 4H), 1.43 (ddd, 6H), 1.96 - 1.12 (m, 4H); LC-
MS: m/z 391.3 (MH+).
Scheme 4, Cpd (III) *i Preparation: 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2-ylpyrazol-3- yl)benzonitrile
Figure imgf000114_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-(6- azaspiro[2.5]octane-6-carbonyl)-3-bromobenzonitrile was used instead of 3-bromo-4-(4-methoxy- 4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 80 %
(130 mg).
NMR (400 MHz, DMSO-d6) δ 8.16 (d, 1H), 7.85 (d, 1H), 7.81 (dd, 1H), 7.45 (d, 1H), 6.60 (d, 1H),
4.60 - 4.45 (m, 1H), 3.76 - 3.49 (m, 2H), 3.15 - 2.85 (m, 2H), 1.50 - 1.40 (m, 7H), 1.37 - 1.30 (m, 1H),
1.22 - 1.12 (m, 1H), 0.93 - 0.85 (m, 1H), 0.40 - 0.13 (m, 4H); LC-MS: 349.2 (MH+).
Scheme 4, Cpd (lll)*i Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-(2-ethylpyrazol-3- yl)benzonitrile
Figure imgf000115_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile and l-ethyl-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole were used instead of 3-bromo-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile and l-(propan-2-yl)-3-(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole respectively. The title compound was obtained in a yield of 30 % (76 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.06 - 7.99 (m, 2H), 7.77 - 7.70 (m, 1H), 7.52 (d, 1H), 6.26 (d,
1H), 4.10 - 3.93 (m, 2H), 3.71 - 3.42 (m, 2H), 3.26 - 2.95 (m, 2H), 2.12 - 1.64 (m, 4H), 1.30 (t, 3H); LC-
MS: m/z 345.24 (MH+).
Scheme 4, Cpd (lll)*k Preparation: 3-[l-(2-methylpropyl)pyrazol-3-yl]-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile
Figure imgf000115_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile and l-(2-methylpropyl)-3-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole (see Scheme 12, Cpd (lll)*e) were used instead of 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile and l-(propan-2-yl)-3-
(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole respectively. The title compound was obtained in a yield of 35 % (12 mg).
1H NMR (400 MHz, DMSO) δ 8.26 - 8.11 (m, 1H), 7.87 - 7.76 (m, 2H), 7.59 - 7.40 (m, 1H), 6.62 (dd,
1H), 4.73 - 4.54 (m, 1H), 3.93 (dd, 2H), 3.27 - 2.53 (m, 4H), 2.11 (dq, 1H), 1.97 - 1.80 (m, 1H), 1.70 -
0.89 (m, 3H), 0.90 - 0.76 (m, 6H); LC-MS: m/z 405.2 (MH+).
Scheme 4, Cpd (lll)*l Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-3- yl)benzonitrile
Figure imgf000116_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile and l-methyl-lH-pyrazole-3-boronic acid pinacol ester were used instead of 3-bromo-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile and l-(propan-2-yl)-3-(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole respectively. The title compound was obtained in a yield of 56 % (56 mg).
1H NMR (400 MHz, DMSO-d6) δ 8.20 (d, 1H), 7.85 (dd, 1H), 7.80 (d, 1H), 7.51 (d, 1H), 6.67 (d, 1H),
3.97 - 3.84 (m, 4H), 3.59 (ddd, 1H), 3.24 - 3.05 (m, 2H), 2.21 - 1.97 (m, 2H), 1.94 - 1.79 (m, 1H),
1.78 - 1.59 (m, 1H); LC-MS: m/z 331.12 (MH+).
Scheme 4, Cpd (lll)*m Preparation: 3-(l-cyclobutylpyrazol-3-yl)-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile
Figure imgf000117_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile and l-cyclobutyl-3-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole (see Scheme 12, Cpd (lll)*a) were used instead of 3- bromo-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile and l-(propan-2-yl)-3-
(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole respectively. The title compound was obtained in a yield of 79 % (165 mg).
1H NMR (400 MHz, DMSO) δ 8.17 (dd, 1H), 7.92 (q, 1H), 7.84 (ddd, 1H), 7.49 (dd, 1H), 6.60 (dd, 1H),
4.84 (p, 1H), 4.62 (d, 1H), 3.21 - 3.11 (m, 1H), 3.03 - 2.69 (m, 2H), 2.57 (d, 1H), 2.44 - 2.30 (m, 3H),
1.97 - 1.65 (m, 4H), 1.56 - 1.12 (m, 3H); LC-MS: m/z 403.2 (MH+).
Scheme 4, Cpd (III) *n Preparation: 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2-ylpyrazol-4- yl)benzonitrile
Figure imgf000117_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile wherein 4-(6- azaspiro[2.5]octane-6-carbonyl)-3-bromobenzonitrile and l-lsopropyl-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole were used instead of 3-bromo-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile and l-(propan-2-yl)-3-(tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole respectively. The title compound was obtained in a yield of 78 % (85 mg).
1H NMR (400 MHz, DMSO) δ 8.08 (d, 1H), 8.03 (dlH), 7.74 (dd, 1H), 7.72 (d, 1H), 7.45 (d, 1H), 4.57
(p, 1H), 3.95 - 3.82 (m, 1H), 3.43 (ddd, 1H), 3.13 - 2.93 (m, 1H), 2.93 - 2.83 (m, 1H), 1.44 (dd, 6H), 1.41 - 1.33 (m, 1H), 1.27 (s, 1H), 0.99 - 0.90 (m, 1H), 0.71 (td, 1H), 0.37 - 0.14 (m, 4H); LC-MS: m/z
349.18 (MH+).
Scheme 4, Cpd (IV)*a Preparation: [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-(4-methoxy-4-methyl-l-piperidyl)methanone
Figure imgf000118_0002
Example 39
A mixture of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile
(120.0 mg, 0.330 mmol), dicesium carbonate (320.08 mg, 0.980 mmol), ethanimidamide hydrochloride (46.44 mg, 0.490 mmol) and CuBr (2.35 mg, 0.020 mmol) in DMSO (3.713 mL) was stirred at 120 °C for 8h. After this time the reaction was cooled down to RT and H2O was added, the mixture was then extracted with AcOEt 3 times. The aqueous phase was acidified until pH = 5 with
HCI IM and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered, concentrated under vacuum and purified by FC on NH column
(from 100% of DCM to DCM/MeOH 95:5) to give the product of formula [2-(l-isopropylpyrazol-3- yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4-methyl-l-piperidyl)methanone.
Yield 63 mg
1H NMR (400 MHz, DMSO) δ 13.78 (s, 1H), 8.33 (dd, 1H), 7.93 (ddd, 1H), 7.82 (dd, 1H), 7.30 (dd,
1H), 6.43 (dd, 1H), 4.55 (dp, 1H), 4.28 - 3.96 (m, 1H), 3.15 - 2.86 (m, 6H), 2.41 (s, 3H), 1.72 (t, 1H),
1.53 - 1.20 (m, 9H), 1.14 - 0.87 (m, 3H); LC-MS: m/z 423.5 (MH+).
Preparation: (4-tert-butyl-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000118_0001
Example 40
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4-tert-butylpiperidine-l-carbonyl)-3-(l- isopropylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4- methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 20 % (22 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.37 (s, 1H), 7.98 - 7.90 (m, 1H), 7.79 (s, 1H), 7.36 - 7.23 (m,
1H), 6.44 - 6.36 (m, 1H), 4.74 - 4.45 (m, 2H), 3.28 - 3.18 (m, 2H), 2.85 - 2.55 (m, 2H), 2.41 (s, 3H),
1.83 - 1.41 (m, 7H), 1.29 - 0.93 (m, 3H), 0.88 - 0.66 (m, 9H), 0.29 - 0.15 (m, 1H); 435.38 (MH+).
Preparation: [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(l-oxa-7- azaspiro[3.5]nonan-7-yl)methanone
Figure imgf000119_0001
Example 41
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 3-(l-isopropylpyrazol-3-yl)-4-(l-oxa-7- azaspiro[3.5]nonane-7-carbonyl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4- methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 50 % (37 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.35 - 8.28 (m, 1H), 7.98 - 7.91 (m, 1H), 7.84 - 7.76 (m, 1H),
7.32 - 7.26 (m, 1H), 6.46 - 6.39 (m, 1H), 4.60 - 4.48 (m, 1H), 4.43 - 4.28 (m, 2H), 3.75 - 3.42 (m, 3H),
3.13 - 2.84 (m, 2H), 2.45 - 2.16 (m, 4H), 1.88 - 1.67 (m, 2H), 1.61 - 1.52 (m, 1H), 1.49 - 1.39 (m, 6H),
1.30 - 1.06 (m, 2H); LC-MS: m/z 421.48 (MH+).
Preparation: (4-ethoxy-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000120_0001
Example 42
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4-ethoxypiperidine-l-carbonyl)-3-(l-isopropylpyrazol-
3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 38 % (80 mg).
1H NMR (400 MHz, DMSO) δ 13.79 (s, 1H), 8.33 (dd, 1H), 7.93 (dt, 1H), 7.81 (dd, 1H), 7.30 (dd, 1H),
6.42 (t, 1H), 4.53 (dp, 1H), 4.05 - 3.91 (m, 1H), 3.48 - 3.36 (m, 2H), 3.28 - 3.05 (m, 3H), 2.95 - 2.74
(m, 1H), 2.41 (s, 3H), 1.79 (d, 1H), 1.55 - 1.32 (m, 8H), 1.29 - 1.18 (m, 1H), 1.06 (dt, 3H); LC-MS: m/z
423.3 (MH+).
Preparation: (4-fluoro-4-methylpiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2- ylpyrazol-3-yl)phenyl]methanone
Figure imgf000120_0002
Example 43
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4-fluoro-4-methylpiperidine-l-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 75 % (32 mg).
1H NMR (400 MHz, DMSO) δ 13.80 (s, 1H), 8.33 (dd, 1H), 7.94 (td, 1H), 7.82 (dd, 1H), 7.33 (t, 1H),
6.43 (dd, 1H), 4.55 (dp, 1H), 4.40 - 4.13 (m, 1H), 3.19 - 2.86 (m, 3H), 2.41 (s, 3H), 1.86 - 1.40 (m,
10H), 1.37 - 1.13 (m, 3H); LC-MS: m/z 411.6 (MH+). Preparation: (4,4-difluoropiperidin-l-yl)-[2-(2-ethylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000121_0001
Example 44
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-(2-ethylpyrazol-3- yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine- l-carbonyl)benzonitrile. The title compound was obtained in a yield of 60 % (53 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.15 - 8.08 (m, 1H), 7.96 (d, 1H), 7.52 (d, 2H), 6.26 (d, 1H), 4.01
(d, 2H), 3.68 - 3.49 (m, 2H), 3.25 - 3.01 (m, 3H), 2.41 (s, 3H), 2.10 - 1.64 (m, 4H), 1.32 (t, 3H); LC-MS: m/z 401.23 (MH+).
Preparation: [2-[l-(2-methylpropyl)pyrazol-3-yl]-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000121_0002
Example 45
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 3-[l-(2-methylpropyl)pyrazol-3-yl]-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-
4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 10 % (4 mg). 1H NMR (400 MHz, DMSO) δ 13.80 (s, 1H), 8.36 (dd, 1H), 7.95 (ddd, 1H), 7.77 (dd, 1H), 7.39 - 7.25
(m, 1H), 6.52 - 6.37 (m, 1H), 4.75 - 4.58 (m, 1H), 3.99 - 3.91 (m, 2H), 3.02 - 2.55 (m, 3H), 2.41 (s, 3H),
2.21 - 2.06 (m, 1H), 1.97 - 1.79 (m, 1H), 1.73 - 0.89 (m, 4H), 0.88 (s, 6H); LC-MS: m/z 461.2 (MH+).
Preparation: (4,4-difluoropiperidin-l-yl)-[2-(l-methylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000122_0001
Example 46
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-methylpyrazol-
3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 36 % (23 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 13.93 - 13.69 (m, 1H), 8.34 (d, 1H), 7.95 (dd, 1H), 7.77 (d, 1H),
7.37 (d, 1H), 6.49 (d, 1H), 3.88 (s, 4H), 3.64 - 3.49 (m, 1H), 3.25 - 3.09 (m, 2H), 2.41 (s, 3H), 2.17 -
1.77 (m, 3H), 1.69 - 1.49 (m, 1H); LC-MS: m/z 387.19 (MH+).
Preparation: [2-(l-cyclobutylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000122_0002
Example 47
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 3-(l-cyclobutylpyrazol-3-yl)-4-[4- (trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-
4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 36 % (67 mg).
1H NMR (400 MHz, DMSO) δ 13.80 (s, 1H), 8.33 (dd, 1H), 7.96 (ddd, 1H), 7.86 (dd, 1H), 7.35 (dd,
1H), 6.46 (t, 1H), 4.87 (p, 1H), 4.66 (d, 1H), 3.40 (d, 1H), 3.21 (d, 1H), 3.03 - 2.70 (m, 2H), 2.56 (d,
1H), 2.47 - 2.31 (m, 6H), 2.03 - 1.63 (m, 3H), 1.56 - 1.18 (m, 2H), 0.40 (qd, 1H); LC-MS: m/z 459.48
(MH+).
Preparation: [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2-ylpyrazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000123_0001
Example 48
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 3-(l-propan-2-ylpyrazol-3-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-
4-(4-methoxy-4-methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 27 % (67 mg).
1H NMR (400 MHz, DMSO) δ 13.80 (s, 1H), 8.43 - 8.23 (m, 1H), 7.95 (ddd, 1H), 7.81 (dd, 1H), 7.39 -
7.27 (m, 1H), 6.42 (d, 1H), 4.66 (d, 1H), 4.54 (pd, 1H), 3.47 - 2.45 (m, 4H), 2.41 (s, 3H), 1.46 (s, 2H),
1.44 (dd, 6H), 1.38 - 0.29 (m, 2H); LC-MS: m/z 447.2 (MH+).
Preparation: 6-azaspiro[2.5]octan-6-yl-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2-ylpyrazol-
4-yl)phenyl]methanone
Figure imgf000124_0001
Example 49 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2- ylpyrazol-4-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 19 % (18 mg).
1H NMR (400 MHz, DMSO) δ 13.77 (s, 1H), 8.09 (d, 1H), 7.97 (s, 1H), 7.88 (dd, 1H), 7.64 (s, 1H), 7.32
(d, 1H), 4.58 (hept, 1H), 3.96 - 3.80 (m, 1H), 3.41 (ddd, 1H), 3.09 - 2.88 (m, 2H), 2.41 (s, 3H), 1.45
(dd, 6H), 1.38 (ddd, 1H), 1.23 (dt, 1H), 0.92 (dt, 1H), 0.71 (ddd, 1H), 0.38 - 0.13 (m, 4H); LC-MS: m/z
405.57 (MH+).
Preparation: 6-azaspiro[2.5]octan-6-yl-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2-ylpyrazol-
3-yl)phenyl]methanone
Figure imgf000124_0002
Example 50
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6-azaspiro[2.5]octane-6-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 39 % (31 mg).
1H NMR (400 MHz, DMSO-d6) δ 13.80 (s, 1H), 8.35 (d, 1H), 7.94 (dd, 1H), 7.84 (d, 1H), 7.32 (d, 1H),
6.46 (d, 1H), 4.57 (p, 1H), 3.77 - 3.54 (m, 2H), 3.04 (dddd, 2H), 2.42 (s, 3H), 1.47 (dd, 6H), 1.35 (q,
2H), 1.16 - 1.06 (m, 1H), 0.85 (ddd, 1H), 0.44 - 0.05 (m, 4H); LC-MS: m/z 405.24 (MH+). Preparation: (4,4-difluoropiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2- ylpyrazol-3-yl)phenyl]methanone
Figure imgf000125_0001
Example 51
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-(l-propan-2- ylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4-methyl- piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 48 % (50 mg).
1H NMR (400 MHz, DMSO-d6 ) δ ppm 14.61 - 12.76 (m, 1H), 8.31 (d, 1H), 7.96 (dd, 1H), 7.84 (d, 1H),
7.38 (d, 1H), 6.48 (d, 1H), 4.54 (spt, 1H), 3.97 - 3.78 (m, 1H), 3.64 - 3.52 (m, 1H), 3.23 - 3.02 (m, 2H),
2.41 (s, 3H), 2.16 - 1.70 (m, 3H), 1.44 (dd, 6H), 1.54 - 1.19 (m, 1H); LC-MS: m/z 415.3 (MH+).
Preparation: (6,6-difluoro-2-azaspiro[3.3]heptan-2-yl)-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H- l,2,4-triazol-3-yl)phenyl]methanone
Figure imgf000125_0002
Example 52
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 39 [2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone wherein 4-(6,6-difluoro-2-azaspiro[3.3]heptane-2-carbonyl)-3-(l- isopropylpyrazol-3-yl)benzonitrile was used instead of 3-(l-isopropylpyrazol-3-yl)-4-(4-methoxy-4- methyl-piperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 35 % (21 mg). 1H NMR (400 MHz, DMS0-d6) δ 13.73 (s, 1H), 8.30 (d, 1H), 7.93 (d, 1H), 7.84 (d, 1H), 7.33 (d, 1H),
6.48 (d, 1H), 4.56 (p, 1H), 4.06 (s, 2H), 3.69 (s, 2H), 2.89 - 2.56 (m, 4H), 2.42 (s, 3H), 1.47 (d, 6H); LC-
MS: m/z 427.30
Scheme 4, Cpd (V)*a Preparation: [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethoxy)piperidin-l-yl]methanone
Figure imgf000126_0001
A mixture of 3-bromo-4-[4-(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile (268.0 mg, 0.710 mmol), dicesium carbonate (694.56 mg, 2.13 mmol), ethanimidamide hydrochloride (100.77 mg,
1.07 mmol) and CuBr (5.1 mg, 0.040 mmol) in DMSO (3.5 mL) was stirred at 120 °C for 7h. After this time the reaction was cooled down, and directly purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O + 0.1% of
HCOOH) affording the product of formula [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethoxy)piperidin-l-yl]methanone.
Yield: 34 mg
1H NMR (400 MHz, DMSO) δ 13.88 (s, 1H), 8.18 (d, 1H), 8.05 - 7.98 (m, 1H), 7.54 - 7.39 (m, 1H), 4.74
(dt, 1H), 4.08 - 3.98 (m, 1H), 3.50 - 3.37 (m, 2H), 3.30 - 3.11 (m, 1H), 2.41 (s, 3H), 2.14 - 1.59 (m, 4H);
LC-MS: m/z 433.07, 435.05 (MH+).
Scheme 4, Cpd (V)*b Preparation: [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4- methylpiperidin-l-yl)methanone
Figure imgf000126_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound [2- bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethoxy)piperidin-l-yl]methanone wherein 3-bromo-4-(4-methylpiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-[4-
(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 61 % (65 mg). 1H NMR (400 MHz, DMSO) δ 13.82 (s, 1H), 8.17 (d, 1H), 8.00 (ddd, 1H), 7.35 (d, 1H), 4.46 (s, 1H),
3.23 (s, 1H), 3.08 - 2.92 (m, 1H), 2.77 (dt, 1H), 2.41 (s, 3H), 1.77 - 1.43 (m, 3H), 1.23 - 0.98 (m, 2H),
0.92 (d, 3H); LC-MS: m/z 363.10, 365.12 (MH+).
Scheme 4, Cpd (V)*c Preparation: [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4- methoxypiperidin-l-yl)methanone
Figure imgf000127_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound [2- bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethoxy)piperidin-l-yl]methanone wherein 3-bromo-4-(4-methoxypiperidine-l-carbonyl)benzonitrile was used instead of 3-bromo-4-
[4-(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 52 % (120 mg).
1H NMR (400 MHz, DMSO) δ 13.86 (s, 1H), 8.17 (d, 1H), 8.00 (dd, 1H), 7.41 (t, 1H), 3.93 (d, 1H), 3.44
(d, 1H), 3.39 - 3.32 (m, 2H), 3.25 (s, 3H), 3.10 - 2.96 (m, 1H), 2.41 (s, 3H), 1.96 - 1.86 (m, 1H), 1.79 (s,
1H), 1.56 - 1.32 (m, 2H); LC-MS: m/z 379.36, 381.36 (MH+).
Scheme 4, Cpd (V)*d Preparation: [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4- (trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000127_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound [2- bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethoxy)piperidin-l-yl]methanone wherein 3-bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3- bromo-4-[4-(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 54 % (252 mg).
1H NMR (400 MHz, DMSO) δ 13.87 (s, 1H), 8.18 (dd, 1H), 8.01 (dt, 1H), 7.51 (dd, 1H), 4.62 (s, 1H),
3.36 (d, 1H), 3.19 - 3.03 (m, 1H), 2.83 (t, 1H), 2.71 - 2.61 (m, 1H), 2.41 (s, 3H), 1.94 (s, 1H), 1.84 - 1.71
(m, 1H), 1.57 - 1.27 (m, 2H); LC-MS: m/z 417.05, 419.07 (MH+). Scheme 4, Cpd (V)*e Preparation: [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone
Figure imgf000128_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound [2- bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethoxy)piperidin-l-yl]methanone wherein 3-bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3- bromo-4-[4-(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 53 % (268 mg).
1H NMR (400 MHz, DMSO-d6) δ 13.88 (s, 1H), 8.20 (d, 1H), 8.03 (dd, 1H), 7.52 (d, 1H), 3.92 - 3.81 (m,
1H), 3.74 - 3.63 (m, 1H), 3.30 - 3.14 (m, 2H), 2.42 (s, 3H), 2.17 - 1.92 (m, 4H); LC-MS: m/z 385.05,
387.06 (MH+).
Preparation: [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone
Figure imgf000128_0001
Example 53
A mixture of tripotassium phosphate (154.67 mg, 0.730 mmol), X-Phos amonobiphenyl palladium chloride precatalyst (15.07 mg, 0.020 mmol), l-tert-butyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole (95 mg, 0.380 mmol) and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone (80.0 mg, 0.190 mmol) in 1,2- dimethoxyethane (2.128 mL)/Water (0.142 mL) was degassed with N2 for 5 min and then by N2/vacuum cycles, 3 times. The reaction was stirred ON at 90 °C. The reaction mixture was cooled down to room temperature and diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The mixture was purified by by FC on NH column (eluting from 100% of
DCM to DCM/MeOH to 95:5) and again by FC on reverse phase using acid conditions (eluting from
5:95 of CH3CN/H2O + 0.1% of HCOOH to 60:40 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone.
Yield: 35 mg
1H NMR (400 MHz, DMSO) δ 13.80 (s, 1H), 8.30 (dd, 1H), 7.95 (ddd, 1H), 7.86 (t, 1H), 7.33 (dd, 1H),
6.60 - 6.26 (m, 1H), 4.65 (d, 1H), 3.45 - 3.16 (m, 1H), 2.93 - 2.67 (m, 2H), 2.49 (d, 1H), 2.41 (s, 3H),
1.97 - 1.63 (m, 1H), 1.55 (d, 9H), 1.47 - 0.09 (m, 3H); LC-MS: m/z 461.3 (MH+).
Preparation: [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone
Figure imgf000129_0001
Example 54
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone was used instead of [2-bromo-4-(5-methyl-4Hl,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone. The title compound was obtained in a yield of 61 % (68 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.29 (d, 1H) 7.92 - 7.98 (m, 1H) 7.89 (d, 1H) 7.34 - 7.43 (m, 1H)
6.44 - 6.50 (m, 1H) 3.67 - 3.77 (m, 2 H) 3.00 - 3.25 (m, 2H) 2.41 (s, 3 H) 1.75 - 2.15 (m, 3 H) 1.55 (s, 9H)
1.25 - 1.37 (m, 2H); LC-MS: m/z 429.29 (MH+).
Preparation: [2-(l-cyclobutylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone
Figure imgf000130_0002
Example 55
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone and l-cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole (see Scheme 12, Cpd (lll)*a) were used instead of [2-bromo-4-(5- methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone and 1-tert- butyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole respectively. The title compound was obtained in a yield of 37 % (29 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 13.91 - 13.70 (m, 1H), 8.31 (d, 1H), 7.99 - 7.94 (m, 1H), 7.88
(d, 1H), 7.39 (d, 1H), 6.51 (d, 1H), 4.87 (p, 1H), 3.82 - 3.68 (m, 2H), 3.26 - 3.09 (m, 2H), 2.48 - 2.35
(m, 6H), 2.15 - 1.75 (m, 6H), 1.56 - 1.38 (m, 1H); LC-MS: m/z 427.27 (MH+).
Preparation: l-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[l-(propan-2-yl)-lH-pyrazol-3-yl]benzoyl]-4-
(trifluoromethoxy)piperidine
Figure imgf000130_0001
Example 56
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-[4-(trifluoromethoxy)piperidin-l-yl]methanone and l-(propan-2-yl)-3-(tetramethyl- l,3,2-dioxaborolan-2-yl)-lH-pyrazole were used instead of [2-bromo-4-(5-methyl-4H-l,2,4-triazol-
3-yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone and l-tert-butyl-3-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyrazole respectively. The title compound was obtained in a yield of 29 %
(10 mg).
1H NMR (400 MHz, DMSO) δ 13.78 (s, 1H), 8.33 (dd, 1H), 7.95 (ddd, 1H), 7.88 - 7.78 (m, 1H), 7.35
(dd, 1H), 6.45 (dd, 1H), 4.69 - 4.49 (m, 2H), 4.15 - 4.00 (m, 1H), 3.26 - 2.90 (m, 3H), 2.42 (s, 3H), 1.96
(d, 1H), 1.80 - 0.82 (m, 9H); LC-MS: m/z 463.5 (MH+).
Preparation: [2-[l-(l-cyclopropylethyl)pyrazol-3-yl]-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoro-l-piperidyl)methanone
Figure imgf000131_0001
Example 57
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone and l-(l-cyclopropylethyl)-3-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole were used instead of [2-bromo-4-(5-methyl-4Hl,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone and l-tert-butyl-3-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole respectively. The title compound was obtained in a yield of 30 % (34 mg) as a racemic mixture.
1H NMR (400 MHz, DMSO-d6) δ 8.32 (t, 1H), 8.00 - 7.93 (m, 1H), 7.87 (t, 1H), 7.39 (d, 1H), 6.48 (dd,
1H), 3.93 - 3.79 (m, 1H), 3.75 - 3.64 (m, 1H), 3.64 - 3.48 (m, 1H), 3.22 - 3.04 (m, 2H), 2.42 (s, 3H),
2.14 - 1.97 (m, 1H), 1.97 - 1.72 (m, 1H), 1.58 - 1.47 (m, 3H), 1.44 - 1.15 (m, 1H), 0.71 - 0.56 (m, 1H),
0.46 - 0.37 (m, 1H), 0.34 (t, 2H); LC-MS: m/z 441.25 (MH+).
The racemic mixture (Example 57) was then separated into single enantiomers (Examples 57a and
57b) by chiral semi-preparative HPLC Column Chiralpak IC (25 x 2.0 cm), 5 μ
Mobile phase n-Hexane/(Ethanol + 0.1% I PA)
Preparative chiral 82/18 % v/v chromatography Flow rate (ml/min) 17 ml/min protocol: DAD detection 220 nm
Loop 1000 μL
Total amount 33 mg
Injection 9.4 mg (each injection)
Enantiomer 1 Rt 19.97 min 100% ee
LC-MS: m/z 441.34 (MH+)
[2-[l-(lR or lS-cyclopropylethyl)pyrazol-3-yl]-4-(5- methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoro-l- piperidyl)methanone
Figure imgf000132_0001
Example 57a
1R or 1S enantiomer
Enantiomer 2 Rt 21.66 min 90% ee
LC-MS: m/z 441.34 (MH+)
[2-[l-(lR or lS-cyclopropylethyl)pyrazol-3-yl]-4-(5- methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoro-l- piperidyl)methanone
Figure imgf000132_0002
Example 57b
1R or 1S enantiomer
Preparation: (4-methoxypiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2-ylpyrazol-
3-yl)phenyl]methanone
Figure imgf000133_0002
Example 58
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-(4-methoxypiperidin-l-yl)methanone and l-(propan-2-yl)-3-(tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole were used instead of [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone and l-tert-butyl-3-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)pyrazole respectively. The title compound was obtained in a yield of 41 %
(55 mg).
1H NMR (400 MHz, DMSO) δ 13.72 (s, 1H), 8.34 (dd, 1H), 7.93 (dt, 1H), 7.81 (dd, 1H), 7.30 (s, 1H),
6.42 (t, 1H), 4.54 (pd, 1H), 4.05 - 3.84 (m, 1H), 3.30 (s, 2H), 3.24 - 3.05 (m, 4H), 2.96 - 2.76 (m, 1H),
2.41 (s, 3H), 1.81 (s, 1H), 1.58 - 0.68 (m, 9H); LC-MS: m/z 409.2 (MH+).
Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-ethylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000133_0001
Example 59
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein l-ethyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone were used instead of l-tert-butyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole (383.71 mg, 0.380 mmol) and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone respectively. The title compound was obtained in a yield of 24 % (12 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.33 (d, 1H), 7.96 (dd, 1H), 7.82 (d, 1H), 7.37 (d, 1H), 6.48 (d,
1H), 4.17 (q, 2H), 3.90 (br d, 1H), 3.56 (m, 1H), 3.24 - 3.08 (m, 2H), 2.41 (s, 3H), 2.17- 1.75 (m, 4H),
1.39 (t, 4H); LC-MS: m/z 401.31 (MH+).
Preparation: (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[l-(oxetan-3- yl)pyrazol-3-yl]phenyl]methanone
Figure imgf000134_0001
Example 60
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein l-(oxetan-3-yl)-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone were used instead of l-tert-butyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone respectively. The title compound was obtained in a yield of 55 % (42 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.36 (d, 1H), 8.02 - 7.91 (m, 2H), 7.42 (d, 1H), 6.60 (d, 1H), 5.68
- 5.58 (m, 1H), 5.00 - 4.84 (m, 4H), 3.86 - 3.65 (m, 2H), 3.23 - 3.11 (m, 2H), 2.45 - 2.35 (m, 3H), 2.17
- 1.83 (m, 4H), 1.67 - 1.50 (m, 1H); LC-MS: m/z 429.38 (MH+).
Preparation: (4-methylpiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(l-propan-2-ylpyrazol-
3-yl)phenyl]methanone
Figure imgf000135_0001
Example 61
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 53 [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone wherein l-(propan-2-yl)-3-(tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4- methylpiperidin-l-yl)methanone were used instead of l-tert-butyl-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole and [2-bromo-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone respectively. The title compound was obtained in a yield of 60 % (22 mg).
1H NMR (400 MHz, DMSO) δ 13.79 (s, 1H), 8.42 - 8.28 (m, 1H), 7.93 (ddd, 1H), 7.81 (dd, 1H), 7.28
(dd, 1H), 6.42 (td, 1H), 4.63 - 4.45 (m, 2H), 3.26 - 3.08 (m, 1H), 2.89 - 2.57 (m, 2H), 2.41 (s, 3H), 1.76
- 1.32 (m, 9H), 1.24 - 0.94 (m, 2H), 0.92 - 0.70 (m, 3H); LC-MS: m/z 393.57 (MH+).
Scheme 5
Figure imgf000135_0002
Step 1
Compound of formula II may be obtained via Miyaura Borylation between compound of formula I
(commercially available from Astatech) and B2Pin2, in the presence of a suitable transition metal catalyst, e.g. Pd(dppf)Cl2, a suitable base, e.g. potassium acetate, in a suitable solvents, e.g. 1,4-
Dioxane, typically at 100 °C. The reaction takes from about 3 hours to about 12 hours to complete.
Step 2
Compound of formula III may be obtained via Suzuki coupling between compound of formula II and the desired heteroaryl halide, in presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 100
°C. The reaction takes from about hours to about 12 hours to complete.
Step 3
Compound of formula IV may be obtained by hydrolysis of compound of formula III with LiOH in presence of a suitable mixture of solvents, such as MeOH/ THF/ water, typically at room temperature. The reaction takes about 12 hours to complete.
Step 4
Compound of formula V may be obtained from compound of formula IV by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
Step 5
Compound of formula VI may be obtained by nitrile cyclization of compound of formula V with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes from about 3 hours to about 12 hours to complete.
Scheme 5, Cpd (II) Preparation: methyl 4-cyano-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)benzoate
Figure imgf000137_0002
A suspension of [l,l'-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (0.07 g, 0.080 mmol), potassium acetate (1.24 g, 12.5 mmol), 4,4,5,5-tetramethyl-
2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (1.27 g, 5 mmol) and methyl 2- bromo-4-cyanobenzoate (1.0 g, 4.17 mmol) in 1,4-Dioxane (30.41 mL) was degassed under nitrogen flux for 10 min and heated to 100 °C overnight. The day after the reaction mixture was cooled down to room temperature and diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum affording the product of formula methyl 4-cyano-2-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)benzoate, which was in the next step without further purifications.
Yield: 1.19 g.
LC-MS: m/z 288.14 (MH+).
Scheme 5, Cpd (lll)*a Preparation: methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate
Figure imgf000137_0001
A mixture of 2-bromo-4-(trifluoromethyl)thiazole (169.7 mg, 0.730 mmol), methyl 4-cyano-2-
(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzoate (210.0 mg, 0.730 mmol), X-Phos Pd G2 (0.06 g, 0.070 mmol) and tripotassium phosphate (0.47 g, 2.19 mmol) in Water (0.336 mL) and 1,2- dimethoxyethane (3.356 mL) was degassed under nitrogen flux for 10 min. and heated to 100 °C for
2h. The reaction mixture was cooled down to room temperature, diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. Crude was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc 90:10) affording the product of formula methyl 4-cyano-
2-[4-(trifluoromethyl)thiazol-2-yl]benzoate.
Yield: 52 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.70 (d, 1H), 8.43 (d, 1H), 8.15 (dd, 1H), 7.91 (d, 1H), 3.72 (s,
3H); LC-MS: m/z 313.09 (MH+).
Scheme 5, Cpd (lll)*b Preparation: methyl 4-cyano-2-(6-cyclopropyl-2-pyridyl)benzoate
Figure imgf000138_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate wherein 2-bromo-6- cyclopropylpyridine was used instead of 2-bromo-4-(trifluoromethyl)thiazole. The title compound was obtained in a yield of 45 % (303.7 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.20 (d, 1H), 7.98 (dd, 1H), 7.81 - 7.76 (m, 2H), 7.59 (dd, 1H),
7.34 (dd, 1H), 3.66 (s, 3H), 2.17 - 2.09 (m, 1H), 0.98 - 0.83 (m, 4H); LC-MS: m/z 279.22 (MH+).
Scheme 5, Cpd (lll)*c Preparation: methyl 2-(l-tert-butylpyrazol-3-yl)-4-cyano-benzoate
Figure imgf000138_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate wherein l-tert-butyl-3-iodopyrazole was used instead of 2-bromo-4-(trifluoromethyl)thiazole. The title compound was obtained in a yield of 12 % (60 mg). 1H NMR (400 MHz, DMSO-d6) δ ppm 8.20 (d, 1H), 7.90 (d, 1H), 7.84 (dd, 1H), 7.65 (d, 1H), 6.73 (d,
1H), 3.77 (s, 3H) 1.53 (s, 9H); LC-MS: m/z 284.20 (MH+).
Scheme 5, Cpd (IV)*a Preparation: 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoic acid
Figure imgf000139_0001
To a solution of methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate (53.0 mg, 0.170 mmol) in THF (1.178 mL) in Methanol (0.294 mL), a solution of lithium hydroxide (8.13 mg, 0.255 mmol) in Water (0.294 mL) was added. The reaction was allowed to stir overnight at room temperature.
After this time the solution was evaporated under reduced pressure and then the residue was taken up with water. Aqueous phase was washed with DCM, acidified until pH = 4 with a IN solution of
HCI and extracted with EtOAc (x6). The combine organic fractions were dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula 4-cyano-2-[4-
(trifluoromethyl)thiazol-2-yl]benzoic acid.
Yield: 44.7 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 13.71 (br s, 1H), 8.68 (s, 1H), 8.30 (d, 1H), 8.12 (dd, 1H), 7.93
(d, 1H); LC-MS: m/z 299.07 (MH+).
Scheme 5, Cpd (IV)*b Preparation: 4-cyano-2-(6-cyclopropyl-2-pyridyl)benzoic acid
Figure imgf000139_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4- cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoic acid wherein methyl 4-cyano-2-(6-cyclopropyl-2- pyridyl)benzoate was used instead of methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate.
The title compound was obtained in a yield of 93 % (269.5 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 13.04 (br s, 1H), 8.13 (d, 1H), 7.94 (dd, 1H), 7.78 - 7.71 (m,
2H), 7.55 (d, 1H), 7.34 (d, 1H), 2.16 - 2.06 (m, 1H), 1.05 - 0.87 (m, 4 H); LC-MS: m/z 265.22 (MH+).
Scheme 5, Cpd (IV)*c Preparation: 2-(l-tert-butylpyrazol-3-yl)-4-cyano-benzoic acid
Figure imgf000140_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4- cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoic acid wherein methyl 2-(l-tert-butylpyrazol-3-yl)-4- cyano-benzoate was used instead of methyl 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoate.
The title compound was obtained in a yield of 100 % (58 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 13.19 (br s, 1H), 8.15 (d, 1H), 7.88 (d, 1H), 7.81 (dd, 1H), 7.62
(d, 1H), 6.65 (d, 1H), 1.55 (s, 9 H); LC-MS: m/z 270.22 (MH+).
Scheme 5, Cpd (V)*a Preparation: 4-(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-
2-yl]benzonitrile
Figure imgf000140_0002
To a stirred solution of 4-cyano-2-[4-(trifluoromethyl)thiazol-2-yl]benzoic acid (44.7 mg, 0.150 mmol) in DMF (1.041 mL) , N,N-Diisopropylethylamine (0.08 mL, 0.450 mmol) and HATU (85.48 mg,
0.220 mmol) were added. The reaction was stirred for 15 minutes at room temperature. Then 4- methylpiperidine (14.86 mg, 0.150 mmol) was added and the reaction mixture was stirred at room temperature overnight. The day after a saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc 70:30) affording the product of formula 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile.
Yield: 46.8 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.71 - 8.66 (m, 1H), 8.45 - 8.34 (m, 1H), 8.07 (dd, 1H), 7.62
(dd, 1H), 4.46 - 4.35 (m, 1H), 3.27 (s, 2H), 3.06 - 2.97 (m, 1H), 2.81 - 2.58 (m, 8 H); LC-MS: m/z 380.20
(MH+).
Scheme 5, Cpd (V)*b Preparation: 3-(6-cyclopropyl-2-pyridyl)-4-(4-methylpiperidine-l- carbonyl)benzonitrile
Figure imgf000141_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 4-cyano-2-
(6-cyclopropyl-2-pyridyl)benzoic acid was used instead of 4-cyano-2-[4-(trifluoromethyl)thiazol-2- yl)benzoic acid. The title compound was obtained in a yield of 85 % (55.3 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.42 - 8.25 (m, 1H), 8.14 - 8.06 (m, 1H), 7.92 (q, 1H), 7.75 -
7.60 (m, 2H), 7.49 (dd, 1H), 4.61 - 4.50 (m, 1H), 2.96 - 2.71 (m, 2H), 2.37 - 2.23 (m, 1H), 1.88 - 1.49
(m, 3H), 1.43 - 1.04 (m, 6H), 0.97 - 0.79 (m, 3H), 0.18 -0.09 (m, 1H); LC-MS: m/z 346.25 (MH+).
Scheme 5, Cpd (V)*c Preparation: 3-(l-tert-butylpyrazol-3-yl)-4-(4-methylpiperidine-l- carbonyl)benzonitrile
Figure imgf000142_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 2-(l-tert- butylpyrazol-3-yl)-4-cyano-benzoic acid was used instead of 4-cyano-2-[4-(trifluoromethyl)thiazol-
2-yl]benzoic acid. The title compound was obtained in a yield of 84 % (60 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.15 (dd, 1H), 7.90 (dd, 1H), 7.81 (ddd, 1H), 7.43 (dd, 1H),
6.56 (dd, 1H), 4.55 - 4.41 (m, 1H), 3.99 - 2.23 (m, 1H), 2.89 - 2.64 (m, 2H), 1.54 (d, 10H), 1.29 - 0.93
(m, 3H), 0.92 - 0.71 (m, 3H), 0.13 (qd, 1H); LC-MS: m/z 351.35 (MH+).
Scheme 5, Cpd (V)*d Preparation: 3-(6-cyclopropyl-2-pyridyl)-4-(4-fluoro-4-methyl-piperidine-l- carbonyl)benzonitrile
Figure imgf000142_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 4-fluoro-4- methyl-piperidine hydrochloride was used instead of 4-methylpiperidine and 4-cyano-2-(6- cyclopropyl-2-pyridyl)benzoic acid was used instead of 4-cyano-2-[4-(trifluoromethyl)thiazol-2- yl)benzoic acid. The title compound was obtained in a yield of 29 % (13 mg)
LC-MS: m/z 364.35 (MH+).
Scheme 5, Cpd (V)*e Preparation: 3-(l-tert-butylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile
Figure imgf000143_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 4- methoxy-4-methyl-piperidine hydrochloride was used instead of 4-methylpiperidine and 2-(l-tert- butylpyrazol-3-yl)-4-cyano-benzoic acid was used instead of 4-cyano-2-[4-(trifluoromethyl)thiazol-
2-yl]benzoic acid. The title compound was obtained in a yield of 81 % (41 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.15 (dd, 1H), 7.90 (dd, 1H), 7.84 - 7.78 (m, 1 ), 7.45 (dd, 1H),
6.58 (dd, 1H), 4.19 - 4.05 (m, 1H), 3.14 - 2.77 (m, 6H), 1.84 - 1.66 (m, 1H), 1.54 (d, 11H), 1.11 - 0.89
(m, 3H), 0.57 - 0.43 (m, 1H); LC-MS: m/z 381.36 (MH+).
Scheme 5, Cpd (V)*f Preparation: 4-(4-methoxy-4-methyl-piperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile
Figure imgf000143_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methylpiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 4- methoxy-4-methyl-piperidine hydrochloride was used instead of 4-methylpiperidine. The title compound was obtained in a yield of 65 % (88.8 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.71 - 8.65 (m, 1H), 8.45 - 8.35 (m, 1H), 8.12 - 8.03 (m, 1H),
7.69 - 7.57 (m, 1H), 4.19 - 4.01 (m, 1H), 3.29 (s, 3H), 3.26 - 2.95 (m, 3H), 1.81 - 1.70 (m, 1H), 1.60 -
1.20 (m, 3H), 1.04 (s, 3H); LC-MS: m/z 410.27 (MH+).
Scheme 5, Cpd (Vl)*a Preparation: (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone
Figure imgf000144_0001
Example 62
A mixture of dicesium carbonate (120.57 mg, 0.370 mmol), ethanimidamide hydrochloride (17.49 mg, 0.190 mmol), CuBr (0.88 mg, 0.010 mmol) and 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile (46.8 mg, 0.120 mmol) in DMSO (1.199 mL) was stirred at 120 °C for 4h. After this time the reaction was cooled down to room temperature and H2O was added. The mixture was then extracted with AcOEt 3 times. Aqueous phase was diluted with a saturated solution of potassium sodium tartrate and extracted with EtOAc and THF x5. The combined organic fractions were dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluenting from 100% of DCM to DCM/MeOH 95:5) affording the product of formula (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl].
Yield: 26.6 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.69 - 8.61 (m, 1H), 8.54 - 8.38 (m, 1H), 8.20 - 8.13 (m, 1H),
7.56 - 7.40 (m, 1H), 4.56 - 4.41 (m, 1H), 3.09 - 2.94 (m, 1H), 3.12 - 2.62 (m, 3H), 2.46 (s, 2H), 1.84 -
0.71 (m, 9H); LC-MS: m/z 436.28 (MH+).
Preparation: [2-(6-cyclopropyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methyl-l- piperidyl)methanone
Figure imgf000144_0002
Example 63 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 62 (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)thiazol-
2-yl]phenyl]methanone wherein 3-(6-cyclopropyl-2-pyridyl)-4-(4-methylpiperidine-l- carbonyl)benzonitrile was used instead of 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 65 % (42 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.23 (br d, 1H), 8.03 (d, 1H), 7.74 (td, 1H), 7.47 - 7.25 (m, 3H),
4.47 - 4.35 (m, 1H), 2.84 - 2.53 (m, 3H), 2.44 - 2.36 (m, 3H), 2.20 - 2.06 (m, 1H), 1.74 - 0.62 (m, 12H),
0.32 - 0.25 (m, 1H); LC-MS: m/z 402.36 (MH+).
Preparation: [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methyl-l- piperidyl)methanone
Figure imgf000145_0001
Example 64
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 62 (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)thiazol-
2-yl]phenyl]methanone wherein 3-(l-tert-butylpyrazol-3-yl)-4-(4-methylpiperidine-l- carbonyl)benzonitrile was used instead of 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 68 % (47.3 mg).
NMR (400 MHz, DMSO-d6) δ ppm 8.30 (dd, 1H), 7.98 - 7.79 (m, 2H), 7.28 (t, 1H), 6.42 (dd, 1H),
4.49 (br d, 1H), 2.85 - 2.57 (m, 2H), 2.40 (s, 3H), 1.55 (d, 11H), 1.30 - 0.67 (m, 7H), 0.11 -0.09 (m,
1H); LC-MS: m/z 407.47 (MH+).
Preparation: [2-(6-cyclopropyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-fluoro-4- methyl-l-piperidyl)methanone
Figure imgf000146_0001
Example 65
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 62 (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)thiazol-
2-yl]phenyl]methanone wherein 3-(6-cyclopropyl-2-pyridyl)-4-(4-fluoro-4-methyl-piperidine-l- carbonyl)benzonitrile was used instead of 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 53 % (8 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.27 - 8.21 (m, 1H), 8.07 - 8.00 (m, 1H), 7.79 - 7.71 (m, 1H),
7.49 - 7.40 (m, 2H), 7.33 - 7.26 (m, 1H), 4.29 - 4.13 (m, 1H), 3.08 - 2.87 (m, 3H), 2.41 (s, 3H), 2.18 -
2.06 (m, 1H), 1.88 - 0.71 (m, 11H), 0.39 - 0.16 (m, 1H); LC-MS: m/z 420.37 (MH+).
Preparation: [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4-methoxy-4- methyl-l-piperidyl)methanone
Figure imgf000146_0002
Example 66
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 62 (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)thiazol-
2-yl]phenyl]methanone wherein 3-(l-tert-butylpyrazol-3-yl)-4-(4-methoxy-4-methyl-piperidine-l- carbonyl)benzonitrile was used instead of 4-(4-methylpiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 40 % (19 mg). 1H NMR (400 MHz, DMS0-d6) δ ppm 8.31 (dd, 1H), 7.98 - 7.85 (m, 2H), 7.30 (dd, 1H), 6.44 (dd, 1H),
4.22 - 4.07 (m, 1H), 3.05 (d, 6H), 2.41 (s, 3H), 1.80 - 1.65 (m, 1H), 1.57 (d, 9H), 1.51 - 1.22 (m, 3H),
1.12 - 0.89 (m, 3H), 0.45 - 0.28 (m, 1H); LC-MS: m/z 437.48 (MH+).
Preparation: (4-methoxy-4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone
Figure imgf000147_0001
Example 67
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 62 (4-methyl-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)thiazol-
2-yl]phenyl]methanone wherein 4-(4-methoxy-4-methyl-piperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile was used instead of 4-(4-methylpiperidine-l-carbonyl)-3-
[4-(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 25 %
(27.3 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.62 (s, 1H), 8.52 - 8.35 (m, 1H), 8.15 (dd, 1H), 7.53 - 7.42 (m,
1H), 4.22 - 3.98 (m, 1H), 3.23 - 2.95 (m, 6H), 2.42 (s, 3H), 1.83 - 1.69 (m, 1H), 1.58 - 1.36 (m, 3H),
1.25 - 0.99 (m, 4H);LC-MS: m/z 466.29 (MH+).
Scheme 6
Figure imgf000148_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Astatech) by coupling with a proper amine in presence of coupling agent, e.g. HATU, and a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
Step 2
Compound of formula III may be obtained via Miyaura Borylation between compound of formula
II and B2Pin2, in the presence of a suitable transition metal catalyst, e.g. Pd(dppf)Cl2, a suitable base, e.g. potassium acetate, in a suitable solvents, e.g. 1,4-Dioxane, typically at 95 °C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula IV may be obtained via Suzuki coupling between compound of formula III and commercially available heteroaryl halide in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base, e.g. Na2CO3, in a suitable mixture of solvents, such as 1,2-dimethoxyethane /water, typically at 95 °C. The reaction takes from about 3 hours to about 12 hours to complete.
Step 4
Compound of formula V may be obtained by nitrile cyclization of compound of formula IV with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g. Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes from about 3 hours to about 12 hours to complete.
Scheme 6, Cpd (ll)*a Preparation: 3-bromo-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile
Figure imgf000149_0001
To a solution of HATU (1.14 g, 2.99 mmol), 2-bromo-4-cyanobenzoic acid (450.0 mg, 1.99 mmol) and 4-(trifluoromethyl)piperidine hydrochloride (415.24 mg, 2.19 mmol) in DMF (9 mL) N,N-
Diisopropylethylamine (2.03 mL, 11.95 mmol) was added. The reaction was stirred at RT for 5 h. After this time the reaction was diluted with H2O and then extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of cHex to
AcOEt/cHex 60:40) affording the product of formula 3-bromo-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile.
Yield: 560 mg
1H NMR (400 MHz, DMSO) δ 8.29 (dd, 1H), 7.95 (ddd, 1H), 7.71 - 7.50 (m, 1H), 4.60 (t, 1H), 3.26 (d,
1H), 3.09 (dtd, 1H), 2.83 (tdd, 1H), 2.66 (d, 1H), 1.94 (t, 1H), 1.82 - 1.65 (m, 1H), 1.55 - 1.29 (m, 2H);
LC-MS: m/z 361.07, 363.10 (MH+).
Scheme 6, Cpd (lll)*a Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)benzonitrile
Figure imgf000149_0002
3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile (950.0 mg, 2.8 mmol), 4, 4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (2.13 g, 8.4 mmol), potassium acetate (832.78 mg, 8.4 mmol) and [1,1'- Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (68.76 mg,
0.080 mmol) were placed in a vial and suspended in 1,4-Dioxane (25 mL). The mixture was degassed through N2-vacuum cycles (3 times). The mixture was stirred under N2 atmosphere at 90
°C overnight. After this time the reaction was cooled down, filtered through a pad of celite and then washed with EtOAc. The organic phase was washed with Brine, dried over Na2SO4 filtered and concentrated under vacuum affording the product of formula 4-(4,4-difluoropiperidine-l-carbonyl)-
3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)benzonitrile which was used in the next step without further purification.
Yield: 1.0 g
LC-MS: m/z 377.26 (MH+).
Scheme 6, Cpd (lll)*b Preparation: 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4-[4- (trifluoromethyl)piperidine-l-carbonyl]benzonitrile
Figure imgf000150_0001
3-bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile (750.0 mg, 2.08 mmol), 4, 4, 5, 5- tetramethyl-2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (1.58 g, 6.23 mmol), potassium acetate (617.7 mg, 6.23 mmol) and [1,1' -
Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (34.0 mg,
0.040 mmol) were suspended in 1,4-Dioxane (20 mL) and the mixture was degassed through N2- vacuum cycles (3 times). The mixture was stirred under N2 atmosphere at 95 0 C overnight. After this time, complete consumption of starting material was observed so the mixture was cooled down to RT and filtered through a pad of celite washing with EtOAc. The organic phase was washed with
Brine, dried over Na2SO4 filtered and concentrated under vacuum affording the product of formula 3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile. The product was used in the next step without further purification.
Yield: 800 mg
LC-MS: m/z 409.16 (MH+). Scheme 6, Cpd (IV)*a Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-(6-isopropyl-2- pyridyl)benzonitrile
Figure imgf000151_0002
Palladium tetrakis triphenylphosphine (80.17 mg, 0.070 mmol) and 2-bromo-6-isopropyl-pyridine
(277.62 mg, 1.39 mmol) were suspended in 1,2-dimethoxyethane (13.88 mL) and then a 2M aqueous solution of Na2CO3 (2.08 mL, 4.16 mmol) was added. The mixture was degassed by 5 cycles of nitrogen/vacuum and 4-(4,4-difluoropiperidine-l-carbonyl)-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)benzonitrile (0.5 g, 1.39 mmol) was added. The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and then was heated to 95 °C for 8h. The reaction mixture was cooled down to room temperature and diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The mixture was purified by FC on RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 60:460 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula 4-(4,4-difluoropiperidine-l-carbonyl)-3-(6-isopropyl-2-pyridyl)benzonitrile.
Yield: 160 mg
NMR (400 MHz, DMSO-d6) δ 8.25 (d, 1H), 7.98 (dd, 1H), 7.89 -7.85 (m, 1H), 7.72 -7.62 (m, 2H),
7.33 (dd, 1H), 4.06 -3.97 (m, 1H), 3.40 -3.24 (m, 2H), 3.14 -3.05 (m, 1H), 2.99 (p, 1H), 2.23 -1.84 (m,
3H), 1.71 -1.51 (m, 1H), 1.29 -1.20 (m, 6H); LC-MS: m/z 370.31 (MH+).
Scheme 6, Cpd (IV)*b Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-[6-
(trifluoromethyl)pyridin-2-yl]benzonitrile
Figure imgf000151_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4,4-difluoropiperidine-l-carbonyl)-3-(6-isopropyl-2-pyridyl)benzonitrile wherein 2-bromo-6-
(trifluoromethyl)pyridine was used instead of 2-bromo-6-isopropyl-pyridine. The title compound was obtained in a yield of 40 % (130 mg).
1H NMR (400 MHz, DMSO) δ 8.37 (d, 1H), 8.26 (t, 1H), 8.20 (d, 1H), 8.06 (dd, 1H), 7.95 (dd, 1H), 7.75
(d, 1H), 4.04 -3.93 (m, 1H), 3.50 -3.39 (m, 1H), 2.54 -2.50 (m, 2H), 2.23 -1.97 (m, 3H), 1.93 -1.83 (m,
1H); LC-MS: m/z 396.26 (MH+).
Scheme 6, Cpd (IV)*c Preparation: 3-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile
Figure imgf000152_0001
Palladium tetrakis triphenylphosphine (42.46 mg, 0.040 mmol) and 3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile (0.33 g, 0.730 mmol) were suspended in 1,2-dimethoxyethane (7.349 mL) and then a 2M aqueous solution of Na3CO3 (1.1 mL, 2.2 mmol) was added. The reaction was degassed by 5 cycles of nitrogen/vacuum and 2-chloro-
4-methyl-6-(trifluoromethyl)pyrimidine (158.9 mg, 0.810 mmol) was added. The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and then was heated to 95 °C for 8h. After this time the reaction mixture was cooled down to room temperature, diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The residue was purified by FC on silica gel (eluting form AcOEt/cHex 5:95 to AcOEt/cHex 60:40) and again by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 40:60 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula 3-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile.
Yield: 34 mg
1H NMR (400 MHz, DMSO) δ 8.60 - 8.42 (m, 1H), 8.10 (dd, 1H), 7.99 (s, 1H), 7.73 - 7.59 (m, 1H), 4.54
(d, 1H), 3.30 (s, 3H), 2.68 (s, 4H), 1.97 (d, 1H), 1.73 (d, 1H), 1.47 (d, 2H); LC-MS: m/z 443.4 (MH+). Scheme 6, Cpd (IV)*d Preparation: 3-(4-cyclopropylthiazol-2-yl)-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile
Figure imgf000153_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile wherein 2-bromo-4-cyclopropylthiazole was used instead of 2-chloro-4- methyl-6-(trifluoromethyl)pyrimidine. The title compound was obtained in a yield of 11 % (33 mg).
1H NMR (400 MHz, DMSO) δ 8.33 - 8.22 (m, 1H), 7.98 (td, 1H), 7.65 - 7.53 (m, 1H), 7.47 (d, 1H), 4.58
(s, 1H), 3.28 (s, 1H), 3.06 - 2.65 (m, 3H), 2.14 - 2.06 (m, 1H), 1.99 - 1.10 (m, 4H), 0.98 - 0.70 (m, 4H);
LC-MS: m/z 406.47 (MH+).
Scheme 6, Cpd (IV)*e Preparation: 3-(6-propan-2-ylpyridin-2-yl)-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile
Figure imgf000153_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile wherein 2-bromo-6-propan-2-ylpyridine was used instead of 2-chloro-4- methyl-6-(trifluoromethyl)pyrimidine. The title compound was obtained in a yield of 37 % (47 mg).
1H NMR (400 MHz, DMSO) δ 8.27 - 8.11 (m, 1H), 7.96 (ddd, 1H), 7.84 (td, 1H), 7.64 (dd, 1H), 7.59 -
7.49 (m, 1H), 7.37 - 7.26 (m, 1H), 4.52 (d, 1H), 3.30 (s, 4H), 2.50 (p, 1H), 1.94 - 0.02 (m, 10H); LC-MS: m/z 402.28 (MH+). Scheme 6, Cpd (IV)*f Preparation: 3-(6-cyclopropylpyridin-2-yl)-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile
Figure imgf000154_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile wherein 2-bromo-6-cyclopropylpyridine was used instead of 2-chloro-4- methyl-6-(trifluoromethyl)pyrimidine. The title compound was obtained in a yield of 34 % (90 mg).
NMR (400 MHz, DMSO-d6) δ 8.25 - 8.09 (m, 1H), 8.01 - 7.86 (m, 1H), 7.82 - 7.72 (m, 1H), 7.64 -
7.46 (m, 2H), 7.35 - 7.25 (m, 1H), 4.54 (d, 1H), 3.12 - 2.59 (m, 3H), 2.11 (tq, 1H), 1.97 - 1.63 (m,
2H), 1.54 - 1.20 (m, 3H), 0.99 - 0.62 (m, 4H); LC-MS: m/z 400.47 (MH+).
Scheme 6, Cpd (IV)*g Preparation: 4-[4-(trifluoromethyl)piperidine-l-carbonyl]-3-[4- (trifluoromethyl)-l,3-thiazol-2-yl]benzonitrile
Figure imgf000154_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile wherein 2-bromo-4-(trifluoromethyl)thiazole was used instead of 2-chloro-4- methyl-6-(trifluoromethyl)pyrimidine. The title compound was obtained in a yield of 23 % (67 mg).
LC-MS: m/z 434.38 (MH+).
Scheme 6, Cpd (IV)*h Preparation: 4-[4-(trifluoromethyl)piperidine-l-carbonyl]-3-[6-
(trifluoromethyl)pyridin-2-yl]benzonitrile
Figure imgf000155_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile wherein 2-bromo-6-(trifluoromethyl)pyridine was used instead of 2-chloro-4- methyl-6-(trifluoromethyl)pyrimidine. The title compound was obtained in a yield of 17 % (77 mg).
LC-MS: m/z 428.10 (MH+).
Scheme 6, Cpd (V)*a Preparation: [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone
Figure imgf000155_0002
Example 68
A mixture of 3-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzonitrile (35.0 mg, 0.080 mmol), dicesium carbonate (77.34 mg, 0.240 mmol), ethanimidamide hydrochloride (11.22 mg, 0.120 mmol) and CuBr (0.57 mg, 0 mmol) in DMSO (0.791 mL) was stirred at 120 °C for 2h. After this time the reaction was cooled down to
RT and H2O was added. The mixture was then extracted with AcOEt 3 times, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of DCM to DCM/MeOH 95:5) affording the product of formula [4-
(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2-yl]phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone.
Yield: 3 mg
1H NMR (400 MHz, MeOD) δ 9.18 - 8.88 (m, 1H), 8.24 (d, 1H), 7.72 (s, 1H), 7.53 (d, 1H), 4.75 (d, 1H),
3.68 (d, 1H), 3.29 - 2.77 (m, 3H), 2.72 (s, 3H), 2.52 (s, 3H), 2.06 (t, 1H), 1.83 (d, 1H), 1.64 (p, 2H); LC-
MS: m/z 499.39 (MH+). Preparation: [2-(4-cyclopropylthiazol-2-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone
Figure imgf000156_0001
Example 69
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 3-(4-cyclopropylthiazol-2-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 48 % (18 mg).
1H NMR (400 MHz, DMSO) δ 13.87 (s, 1H), 8.34 (dd, 1), 8.12 - 8.03 (m, 1H), 7.56 - 7.29 (m, 2H), 4.62
(s, 1H), 3.41 (d, 1H), 3.07 - 2.57 (m, 3H), 2.42 (s, 3H), 2.11 (tt, 1H), 2.02 - 1.84 (m, 1H), 1.74 - 1.53
(m, 1H), 1.52 - 1.01 (m, 2H), 0.99 - 0.66 (m, 4H); LC-MS: m/z 462.22 (MH+).
Preparation: [2-(6-isopropyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)-l-piperidyl]methanone
Figure imgf000156_0002
Example 70
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 3-(6-propan-2-ylpyridin-2-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 47 % (25 mg). 1H NMR (400 MHz, DMSO) δ 13.83 (s, 1H), 8.31 - 8.25 (m, 1H), 8.10 - 8.02 (m, 1H), 7.82 (t, 1H), 7.58
- 7.39 (m, 2H), 7.34 - 7.23 (m, 1H), 4.55 (d, 1H), 3.61 - 2.55 (m, 4H), 2.41 (s, 4H), 1.97 - 1.33 (m, 3H),
1.30 - 1.20 (m, 6H), 0.98 - 0.13 (m, 1H); LC-MS: m/z 458.48 (MH+).
Preparation: [2-(6-cyclopropylpyridin-2-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000157_0001
Example 71
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 3-(6-cyclopropylpyridin-2-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 17 % (17 mg).
1H NMR (400 MHz, DMSO-d6) δ 13.83 (s, 1H), 8.30 - 8.15 (m, 1H), 8.06 (dt, 1H), 7.76 (td, 1H), 7.51 -
7.35 (m, 2H), 7.28 (dd, 1H), 4.57 (d, 1H), 4.24 - 3.43 (m, 1H), 3.22 - 2.58 (m, 3H), 2.42 (s, 5H), 2.00 -
1.63 (m, 1H), 1.55 - 0.19 (m, 6H); LC-MS: m/z 456.22 (MH+).
Preparation: [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-(trifluoromethyl)-l,3-thiazol-2-yl]phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000157_0002
Example 72
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 4-[4-(trifluoromethyl)piperidine-l- carbonyl]-3-[4-(trifluoromethyl)-l,3-thiazol-2-yl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 48 % (37 mg).
1H NMR (400 MHz, DMSO) δ 13.91 (s, 1H), 8.65 (d, 1H), 8.44 (d, 1H), 8.18 (dd, 1H), 7.53 (dd, 1H),
4.62 (d, 1H), 3.49 (d, 1H), 3.12 (t, 1H), 2.80 (t, 1H), 2.60 (s, 1H), 2.44 (s, 3H), 2.05 - 1.29 (m, 3H), 1.03
(d, 1H); LC-MS: m/z 490.13 (MH+).
Preparation: [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[6-(trifluoromethyl)pyridin-2-yl]phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000158_0001
Example 73
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 4-[4-(trifluoromethyl)piperidine-l- carbonyl]-3-[6-(trifluoromethyl)pyridin-2-yl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 36 % (31 mg).
1H NMR (400 MHz, DMSO) δ 13.86 (s, 1H), 8.32 (d, 1H), 8.23 (t, 1H), 8.13 (dd, 1H), 8.06 (d, 1H), 7.92
(s, 1H), 7.52 (d, 1H), 4.51 (d, 1H), 3.50 (d, 1H), 3.10 - 2.70 (m, 2H), 2.59 (s, 1H), 2.43 (s, 3H), 2.00 -
1.37 (m, 3H), 0.77 (d, 1H); LC-MS: m/z 484.13 (MH+).
Preparation: 4,4-difluoro-l-piperidyl)-[2-(6-isopropyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]methanone
Figure imgf000158_0002
Example 74
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 4-(4,4-difluoropiperidine-l- carbonyl)-3-(6-isopropyl-2-pyridyl)benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 50 % (80 mg).
NMR (400 MHz, DMSO-d6) δ 13.75 (bs, 1H), 8.30 (d, 1H), 8.07 (dd, 1H), 7.85 (t, 1H), 7.54 (dd, 2H),
7.30 (d, 1H), 3.93 -3.83 (m, 1H), 3.45 -3.37 (m, 2H), 3.15 -3.07 (m, 1H), 3.01 (p, 1H), 2.42 (s, 3H), 2.16
-1.74 (m, 3H), 1.56 -1.44 (m, 1H), 1.25 (dd, 6H); LC-MS: m/z 426.35 (MH+).
Preparation: (4,4-difluoropiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[6-
(trifluoromethyl)pyridin-2-yl]phenyl]methanone
Figure imgf000159_0001
Example 75
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 68 [4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-methyl-6-(trifluoromethyl)pyrimidin-2- yl]phenyl]-[4-(trifluoromethyl)-l-piperidyl]methanone wherein 4-(4,4-difluoropiperidine-l- carbonyl)-3-[6-(trifluoromethyl)pyridin-2-yl]benzonitrile was used instead of 3-[4-methyl-6-
(trifluoromethyl)pyrimidin-2-yl]-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of 26 % (25 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.33 (d, 1H), 8.28 - 8.20 (m, 1H), 8.17 - 8.08 (m, 2H), 7.92 (d,
1H), 7.59 (d, 1H), 3.96 - 3.83 (m, 2H), 3.58 - 3.34 (m, 3H), 2.42 (s, 3H), 2.18 - 1.71 (m, 4H); LC-MS: m/z 452.20 (MH+).
Scheme 7
Figure imgf000160_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Astatech) by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 3 hours to about 12 hours to complete.
Step 2
Compound of formula III may be obtained via Miyaura Borylation between compound of formula II
(commercially available from Astatech) and 2-(5,5-dimethyl-l,3,2-dioxaborinan-2-yl)-5,5-dimethyl-
1,3,2-dioxaborinane, in the presence of a suitable transition metal catalyst, e.g. Pd(dppf)Cl2, a suitable base, e.g. potassium acetate, in a suitable solvents, e.g. 1,4-Dioxane, typically at 95 °C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula IV may be obtained via Suzuki coupling between compound of formula III and commercially available heteroaryl halide in the presence of a suitable palladium catalyst, e.g. palladium tetrakis triphenylphosphine, suitable inorganic base Na2CO3, in a suitable mixture of solvents, 1,2-dimethoxyethane /water, typically at 95 °C. The reaction takes from about 3 hours to about 12 hours to complete.
Step 4
Compound of formula V may be obtained by nitrile cyclization of compound of formula IV with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g. Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Scheme 7, Cpd (ll)*a Preparation: 3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile
Figure imgf000161_0002
To a solution of [dimethylamino(3-triazolo[4,5-b]pyridinyloxy)methylidene]-dimethylammonium hexafluorophosphate HATU (1.14 g, 2.99 mmol), 2-bromo-4-cyanobenzoic acid (450.0 mg, 1.99 mmol) and N,N-Diisopropylethylamine (2.03 mL, 11.95 mmol) in DMF (13.27 mL), 4,4- difluoropiperidine hydrochloride (376.49 mg, 2.39 mmol) was added. The reaction was stirred at RT for 3h. After this time a saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from 100% of cHex to cHexAcOEt/ 65:35) affording the product of formula 3-bromo-4-(4,4-difluoropiperidine-l- carbonyl)benzonitrile.
Yield: 500 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.31 (d, 1H) 7.98 (dd, 1H) 7.66 (d, 1H) 3.82 - 3.93 (m, 1H) 3.60
- 3.73 (m, 1H) 3.22 (t, 2H) 1.88 - 2.22 (m, 4H); LC-MS: m/z 329.00, 331.00 (MH+).
Scheme 7. Cpd (ll)*b Preparation: 3-bromo-4-[4-(trifluoromethoxy)piperidine-l- carbonyl)benzonitrile
Figure imgf000161_0001
To a solution of [dimethylamino(3-triazolo[4,5-b]pyridinyloxy)methylidene]-dimethylammonium hexafluorophosphate HATU (630.83 mg, 1.66 mmol), 2-bromo-4-cyanobenzoic acid (250.0 mg, 1.11 mmol) and 4-(trifluoromethoxy)piperidine hydrochloride (272.9 mg, 1.33 mmol) in DMF (5.821 mL) N,N-Diisopropylethylamine (1.13 mL, 6.64 mmol) was added. The reaction was stirred at RT for
6 h. After this time the reaction was diluted with H2O. The mixture was then extracted with AcOEt
3 times. The combined organic fractions were washed with brine, dried over a Na2SO4 filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from AcOEtxHex from 5:95 to 60:40) affording the product of formula 3-bromo-4-[4-(trifluoromethoxy)piperidine-l- carbonyl]benzonitrile.
Yield: 260 mg
1H NMR (400 MHz, DMSO) δ 8.29 (dd, 1H), 7.96 (ddd, 1H), 7.72 - 7.49 (m, 1H), 4.74 (dt, 1H), 4.02
(td, 1H), 3.42 (dddd, 1H), 3.30 - 3.06 (m, 2H), 2.09 - 1.97 (m, 1H), 1.90 (s, 1H), 1.84 - 1.56 (m, 2H);
LC-MS: m/z 377.36, 379.26 (MH+).
Scheme 7, Cpd (lll)*a Preparation: [5-cyano-2-(4,4-difluoropiperidine-l-carbonyl)phenyl]boronic acid
Figure imgf000162_0002
Potassium acetate (40.67 mg, 0.410 mmol), [1,1'-
Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (5.6 mg,
0.010 mmol), 3-bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile (45.0 mg, 0.140 mmol) and 2-(5,5-dimethyl-l,3,2-dioxaborinan-2-yl)-5,5-dimethyl-l,3,2-dioxaborinane (46.33 mg,
0.210 mmol) were suspended in 1,4-Dioxane (1 mL). The mixture was then degassed through N2- vacuum cycles 3 times. The vial was sealed and the mixture was stirred at 90 °C for 3h. After this time the reaction was cooled down, filtered through a pad of celite washing with EtOAc. The organic phase was washed with a s.s. of NaHCO3, brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula [5-cyano-2-(4,4-difluoropiperidine-l- carbonyl)phenyl]boronic acid. The product was used in the next step without further purification.
Yield: 40 mg
LC-MS: m/z 295.24 (MH+).
Scheme 7, Cpd (lll)*b Preparation: [5-cyano-2-[4-(trifluoromethoxy)piperidine-l- carbonyl]phenyl]boronic acid
Figure imgf000162_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound [5- cyano-2-(4,4-difluoropiperidine-l-carbonyl)phenyl]boronic acid wherein 3-bromo-4-[4-
(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile was used instead of 3-bromo-4-(4,4- difluoropiperidine-l-carbonyl)benzonitrile. The title compound was obtained in a yield of 61 % (145 mg).
LC-MS: m/z 343.20 (MH+).
Scheme 7, Cpd (IV)*a Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile
Figure imgf000163_0001
Palladium tetrakis triphenylphosphine (15.72 mg, 0.010 mmol) and [5-cyano-2-(4,4- difluoropiperidine-l-carbonyl)phenyl]boronic acid (80.0 mg, 0.270 mmol) were suspended in 1,2- dimethoxyethane (2 mL) and then a 2 M aqueous solution of Na2CO3 (86.5 mg, 0.820 mmol) was added. The mixture was degassed by 5 cycles of nitrogen/vacuum and then 2-bromo-4-
(trifluoromethyl)thiazole (69.43 mg, 0.300 mmol). The reaction mixture was degassed again by 5 cycles of nitrogen/vacuum and heated to 100 °C overnight. After this time the reaction was cooled down to room temperature, diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The mixture was purified by FC on silica gel (eluting from 100% of cHex to cHex:EtOAc 50:50) to give the product of formula 4-(4,4-difluoropiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)thiazol-2-yl]benzonitrile.
Yield: 35 mg
1H NMR (400 MHz, DMSO-d6) δ 8.69 (d, 1H), 8.44 (d, 1H), 8.11 (dd, 1H), 7.70 (d, 1H), 3.87 - 3.78 (m,
1H), 3.63 - 3.53 (m, 1H), 2.20 - 1.85 (m, 6H); LC-MS: m/z 402.15 (MH+).
Scheme 7, Cpd (IV)*b Preparation: 4-(4,4-difluoropiperidine-l-carbonyl)-3-[4-
(trifluoromethyl)pyrimidin-2-yl]benzonitrile
Figure imgf000164_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4,4-difluoropiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 2- chloro-4-(trifluoromethyl)pyrimidine was used instead of 2-bromo-4-(trifluoromethyl)thiazole. The title compound was obtained in a yield of 40 % (44 mg).
1H NMR (400 MHz, DMSO-d6) δ 9.33 (d, 1H), 8.58 (d, 1H), 8.15 (dd, 1H), 8.09 - 8.04 (m, 1H), 7.76
(dd, 1H), 4.17 - 3.86 (m, 1H), 2.21 - 1.92 (m, 6H); LC-MS: m/z 397.21 (MH+).
Scheme 7, Cpd (IV)*c Preparation: 3-(6-cyclopropyl-2-pyridyl)-4-(4,4-difluoropiperidine-l- carbonyl)benzonitrile
Figure imgf000164_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4,4-difluoropiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 2- bromo-6-cyclopropylpyridine was used instead of 2-bromo-4-(trifluoromethyl)thiazole. The title compound was obtained in a yield of 47 % (28 mg).
1H NMR (400 MHz, DMSO) δ 8.22 (d, 1H), 7.96 (td, 2H), 7.79 (t, 1H), 7.67 (dd, 2H), 3.93 (d, 1H), 3.74
(s, 1H), 3.41 (t, 1H), 2.98 (d, 1H), 2.20 - 1.81 (m, 6H), 1.41 (s, 1H), 1.00 - 0.91 (m, 2H); LC-MS: m/z
368.22 (MH+).
Scheme 7, Cpd (IV)*d Preparation: 3-(6-tert-butyl-2-pyridyl)-4-(4,4-difluoropiperidine-l- carbonyl)benzonitrile
Figure imgf000165_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4,4-difluoropiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein 2- bromo-6-tert-butyl-pyridine was used instead of 2-bromo-4-(trifluoromethyl)thiazole. The title compound was obtained in a yield of 82% (94mg).
H NMR (400 MHz, DMSO) δ 8.22 (d, 1H), 7.99 (dd, 1H), 7.87 (t, 1H), 7.73 (d, 1H), 7.61 (dd, 1H), 7.47
(dd, 1H), 4.11 - 3.96 (m, 1H), 3.25 - 3.14 (m, 2H), 3.06 - 2.90 (m, 1H), 2.19 - 1.77 (m, 3H), 1.56 - 1.38
(m, 1H), 1.30 (s, 9H); LC-MS: m/z 384.47 (MH+).
Scheme 7, Cpd (IV)*e Preparation: 3-(l-tert-butylpyrazol-3-yl)-4-[4-(trifluoromethoxy)piperidine- l-carbonyl]benzonitrile
Figure imgf000165_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4,4-difluoropiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile wherein [5- cyano-2-[4-(trifluoromethoxy)piperidine-l-carbonyl]phenyl]boronic acid and l-tert-butyl-3- iodopyrazole were used instead of [5-cyano-2-(4,4-difluoropiperidine-l-carbonyl)phenyl]boronic acid and 2-bromo-4-(trifluoromethyl)thiazole respectively. The title compound was obtained in a yield of 33% (96 mg).
1H NMR (400 MHz, DMSO) δ 8.22 - 8.12 (m, 1H), 7.96 - 7.88 (m, 1H), 7.82 (td, 1H), 7.50 (dd, 1H),
6.61 (dd, 1H), 4.65 (dt, 1H), 3.51 (ddd, 1H), 3.28 - 2.88 (m, 3H), 1.99 (s, 2H), 1.81 - 1.63 (m, 2H), 1.54
(d, 9H); LC-MS: m/z 421.23 (MH+). Scheme 7, Cpd (V)*a Preparation: (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-
[4-(trifluoromethyl)thiazol-2-yl]phenyl]methanone
Figure imgf000166_0002
Example 76
A mixture of 4-(4,4-difluoropiperidine-l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile
(35.0 mg, 0.070 mmol),CuBr (0.52 mg, 0 mmol), dicesium carbonate (94.33 mg, 0.290 mmol) and ethanimidamide hydrochloride (13.69 mg, 0.140 mmol) in DMSO (1.334 mL) was stirred at 120
°C for 2h. After this time the reaction was cooled down to RT. H2O was added and the mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with Brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column
(eluting from 100% of DCM to DCM:MeOH 95:5) and again by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 55:45 of CH3CN/H2O + 0.1% of
HCOOH) affording the product of formula (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3- yl)-2-[4-(trifluoromethyl)thiazol-2-yl]phenyl]methanone.
Yield: 14 mg
1H NMR (400 MHz, DMSO-d6) δ 8.65 (d, 1H), 8.43 (d, 1H), 8.19 (dd, 1H), 7.56 (d, 1H), 3.94 - 3.82 (m,
1H), 3.67 - 3.52 (m, 1H), 3.38 - 3.30 (m, 2H), 2.44 (s, 3H), 2.20 - 1.79 (m, 4H); LC-MS: m/z 458.38
(MH+).
Preparation: (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)pyrimidin-2-yl]phenyl]methanone
Figure imgf000166_0001
Example 77
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 76 (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone wherein 4-(4,4-difluoropiperidine-l-carbonyl)-3-
[4-(trifluoromethyl)pyrimidin-2-yl]benzonitrile was used instead of 4-(4,4-difluoropiperidine-l- carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 11% (5 mg).
1H NMR (400 MHz, DMSO-d6) δ 9.30 (d, 1H), 8.86 (d, 1H), 8.19 (dd, 1H), 8.01 (d, 1H), 7.56 (d, 1H),
3.95 - 3.82 (m, 1H), 3.58 - 3.40 (m, 2H) 2.55 - 2.52 (m, 2H), 2.43 (s, 3H), 2.20 - 1.90 (m, 4H); LC-MS: m/z 453.26 (MH+).
Preparation: [2-(6-cyclopropyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoro-l- piperidyl)methanone
Figure imgf000167_0001
Example 78
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 76 (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone wherein 3-(6-cyclopropyl-2-pyridyl)-4-(4,4- difluoropiperidine-l-carbonyl)benzonitrile was used instead of 4-(4,4-difluoropiperidine-l- carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 33% (8 mg).
1H NMR (400 MHz, DMSO-d6) δ 13.80 (bs, 1H), 8.26 (d, 1H), 8.06 (dd, 1H), 7.78 (t, 1H), 7.56 - 7.46
(m, 2H), 7.30 (dd, 1H), 3.92 - 3.80 (m, 1H), 3.54 - 3.45 (m, 2H), 3.08 - 2.93 (m, 1H), 2.42 (s, 3H),
2.19 - 1.80 (m, 3H), 1.36 (s, 1H), 1.12 - 0.69 (m, 4H); LC-MS: m/z 424.31 (MH+).
Preparation: [2-(6-tert-butyl-2-pyridyl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoro-l- piperidyl)methanone
Figure imgf000168_0001
Example 79
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 76 (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone wherein 3-(6-tert-butyl-2-pyridyl)-4-(4,4- difluoropiperidine-l-carbonyl)benzonitrile was used instead of 4-(4,4-difluoropiperidine-l- carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile. The title compound was obtained in a yield of 35% (38 mg).
1H NMR (400 MHz, DMSO) δ 13.85 (s, 1H), 8.29 (d, 1H), 8.07 (dd, 1H), 7.86 (t, 1H), 7.62 - 7.49 (m,
2H), 7.45 (dd, 1H), 3.93 (d, 1H), 3.41 (s, 2H), 3.01 (t, 1H), 2.42 (s, 3H), 1.93 (d, 3H), 1.32 (s, 10H); LC-
MS: m/z 440.31 (MH+)
Preparation [2-(l-tert-butylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethoxy)-l-piperidyl]methanone
Figure imgf000168_0002
Example 80
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 76 (4,4-difluoro-l-piperidyl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-[4-
(trifluoromethyl)thiazol-2-yl]phenyl]methanone wherein 3-(l-tert-butylpyrazol-3-yl)-4-[4-
(trifluoromethoxy)piperidine-l-carbonyl]benzonitrile was used instead of 4-(4,4-difluoropiperidine- l-carbonyl)-3-[4-(trifluoromethyl)thiazol-2-yl]benzonitrile . The title compound was obtained in a yield of 3.5% (4 mg). 1H NMR (400 MHz, DMSO) δ 13.73 (s, 1H), 8.30 (dd, 1H), 8.01 - 7.81 (m, 2H), 7.33 (d, 1H), 6.53 -
6.43 (m, 1H), 4.67 - 4.55 (m, 1H), 4.14 - 3.95 (m, 1H), 3.29 - 2.85 (m, 3H), 2.42 (s, 3H), 1.99 - 1.88 (m,
1H), 1.74 - 1.61 (m, 1H), 1.56 (d, 11H); LC-MS: m/z 477.49 (MH+).
Scheme 8
Figure imgf000169_0001
Step 1
Compound of formula II may be obtained from compound of formula I (synthesized according to the Scheme 7) via Suzuki coupling with 2-bromo-4-thiazolecarboxylic acid ethyl ester, in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 90 °C. The reaction takes about 5 hours to complete.
Step 2
Compound of formula III may be obtained by nitrile cyclization of compound of formula II with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about from 3 hours to about 12 hours to complete.
Step 3
Compound of formula IV may be obtained from compound of formula III by hydrolysis, e.g. using
LiOH, in a suitable mixture of solvents such as MeOH/THF/H2O, typically at 50 °C. The reaction takes about 12 hours to complete.
Scheme 8, Cpd (ll) Preparation: ethyl 2-[5-cyano-2-(4,4-difluoropiperidine-l- carbonyl)phenyl]thiazole-4-carboxylate
Figure imgf000170_0001
Example 81
In a proper vial 2-bromo-4-thiazolecarboxylic acid ethyl ester (134.88 mg, 0.570 mmol), [5-cyano-2-
(4,4-difluoropiperidine-l-carbonyl)phenyl]boronic acid (560.0 mg, 0.380 mmol), X-phos aminobiphenyl palladium chloride precatalyst (29.93 mg, 0.040 mmol), tripotassium phosphate
(242.54 mg, 1.14 mmol) were suspended in a mixture of Water (0.6 mL), 1,2-dimethoxyethane (5.7 mL). The resulting suspension was degassed with nitrogen and heated to 90 °C for 5h. After this time the reaction mixture was cooled down to room temperature, diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. Crude was purified by FC on silica gel
(eluting from cHex/EtOAc 95:5 to cHex/EtOAc 50:50) affording the product of formula ethyl 2-[5- cyano-2-(4,4-difluoropiperidine-l-carbonyl)phenyl]thiazole-4-carboxylate.
Yield:115 mg
1H NMR (400 MHz, DMSO) δ 8.70 (s, 1H), 8.42 (d, 1H), 8.09 (dd, 1H), 7.68 (d, 1H), 4.40 - 4.26 (m,
2H), 3.97 - 3.83 (m, 1H), 3.68 - 3.52 (m, 1H), 3.43 - 3.33 (m, 2H), 2.29 - 1.86 (m, 4H), 1.32 (t, 3H); LC-
MS: m/z 406.37 (MH+).
Scheme 8, Cpd (III) Preparation: ethyl 2-[2-(4,4-difluoropiperidine-l-carbonyl)-5-(5-methyl-4H- l,2,4-triazol-3-yl)phenyl]thiazole-4-carboxylate
Figure imgf000170_0002
Example 82
A mixture of ethyl 2-[5-cyano-2-(4,4-difluoropiperidine-l-carbonyl)phenyl]thiazole-4-carboxylate
(115.0 mg, 0.280 mmol), CuBr (2.03 mg, 0.010 mmol), dicesium carbonate (277.26 mg, 0.850 mmol) and ethanimidamide hydrochloride (40.23 mg, 0.430 mmol) in DMSO (3.618 mL) was stirred at 120
°C for 90 min. After this time the reaction was cooled down to RT and H2O was added. The mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with Brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of DCM to DCM:EtOH 90:10) affording the product of formula ethyl 2-[2-
(4,4-difluoropiperidine-l-carbonyl)-5-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]thiazole-4- carboxylate.
Yield: 30 mg
1H NMR (400 MHz, DMSO-d6) δ 13.90 (bs, 1H), 8.66 (s, 1H), 8.42 (d, 1H), 8.16 (dd, 1H), 7.54 (d, 1H),
4.34 (q, 2H), 4.01 - 3.92 (m, 1H), 3.65 - 3.55 (m, 1H), 3.41 - 3.36 (m, 2H), 2.44 (s, 3H), 2.29 - 1.83 (m,
4H), 1.33 (t, 3H); LC-MS: m/z 462.38 (MH+).
Scheme 8, Cpd (IV) Preparation: 2-[2-(4,4-difluoropiperidine-l-carbonyl)-5-(5-methyl-4H-l,2,4- triazol-3-yl)phenyl]thiazole-4-carboxylic acid
Figure imgf000171_0001
Figure imgf000171_0002
Example 83
To a solution of ethyl 2-[2-(4,4-difluoropiperidine-l-carbonyl)-5-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]thiazole-4-carboxylate (23.0 mg, 0.040 mmol) in Methanol (0.142 mL)/THF (0.712 mL) a solution of lithium hydroxide (1.61 mg, 0.070 mmol) in Water (0.142 mL) was added. The reaction was stirred at 50 °C ON. The day after the reaction was cooled down to RT, acidified with HCI 1 N until pH = 1 and directly purified by FC on reverse phase using acid conditions (eluting from
CH3CN/H2O + 0.1% of FA 5:95 + 0.1 % of FA to CH3CN/H2O + 0.1% of FA 35:65) affording the product of formula 2-[2-(4,4-difluoropiperidine-l-carbonyl)-5-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]thiazole-4-carboxylic acid.
Yield: 6 mg
1H NMR (400 MHz, DMSO) δ 13.86 (s, 1H), 8.71 - 8.30 (m, 2H), 8.14 (dd, 1H), 7.50 (d, 1H), 4.17 -
4.03 (m, 1H), 3.52 - 3.29 (m, 3H), 2.45 (s, 3H), 2.31 - 1.78 (m, 4H).
Scheme 9
Figure imgf000172_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Enamine) by coupling with 4,4-difluoropiperidine hydrochloride in the presence of a suitable coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes about 12 hours to complete.
Step 2
Compound of formula III may be obtained via Suzuki coupling between compound of formula II and l-(l-Methylethyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-1H-pyrazole (commercially available from Enamine), in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2-dimethoxyethane / water, typically at 95 °C.
The reaction takes about 3 hours to complete.
Step 3
Compound of formula IV may be obtained via Stille coupling reaction between compound of formula III and a proper THP protected stannane, in the presence of a suitable palladium precatalyst, e.g. palladium tritert-butylphosphine, a suitable base, e.g. CsF, in a suitable solvent, e.g.
1,4-dioxane, typically at 100 °C. The reaction takes from about 3 hours to about 5 hours to complete.
Step 4 Compound V may be obtained from compound of formula IV by removing the THP protecting group under acidic conditions, e.g. HCI 6N solution, typically at 50 °C. The reaction takes about 3 hours to complete.
Scheme 9, Cpd (II) Preparation: (2-bromo-4-chlorophenyl)-(4,4-difluoropiperidin-l-yl)methanone
Figure imgf000173_0001
To a solution of HATU (1.45 g, 3.82 mmol), 2-bromo-4-chlorobenzoic acid (600.0 mg, 2.55 mmol) and N,N-Diisopropylethylamine (2.66 mL, 15.29 mmol) in DMF (16.99 mL), 4,4- difluoropiperidine;hydrochloride (481.89 mg, 3.06 mmol) was added. The reaction was stirred at RT overnight. The day after a saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified by FC on silica gel (eluting from 100% of cHex to
AcOEt/cHex 30:70) affording the product of formula (2-bromo-4-chlorophenyl)-(4,4- difluoropiperidin-l-yl)methanone.
Yield: 715 mg
1H NMR (400 MHz, DMSO) δ 7.87 (d, 1H), 7.58 (dd, 1H), 7.48 (d, 1H), 3.86 (dt, 1H), 3.66 (ddd, 1H),
3.24 (t, 2H), 2.31 - 1.85 (m, 4H); LC-MS: m/z 338.00, 340.03 (MH+).
Scheme 9, Cpd (III) Preparation: [4-chloro-2-(l-propan-2-ylpyrazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone
Figure imgf000173_0002
In a proper vial, a mixture of X-phos aminobiphenyl palladium chloride precatalyst (162.46 mg,
0.210 mmol), tripotassium phosphate (1316.58 mg, 6.2 mmol), (2-bromo-4-chlorophenyl)-(4,4- difluoropiperidin-l-yl)methanone (700.0 mg, 2.07 mmol) and l-(l-Methylethyl)-3-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (390.53 mg, 1.65 mmol) in Water (0.952 mL)/l,2- dimethoxyethane (14.95 mL) was degassed bubbling N2 for 10 min. The mixture was then subjected to N2-vacuum cycles and then stirred at 95 °C for 3h. After this time the reaction was cooled down to RT. Water was added to the mixture and aqueous phase was extracted with AcOEt 3 times.
Organic phases were combined, washed with brine, dried over Na2SO4 and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc
40:60) affording the product of formula [4-chloro-2-(l-propan-2-ylpyrazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone.
Yield: 450 mg,
1H NMR (400 MHz, DMSO-d6) δ 7.84 (d, 1H), 7.76 (d, 1H), 7.45 (dd, 1H), 7.34 (d, 1H), 6.55 (d, 1H),
4.51 (hept, 1H), 3.82 - 3.61 (m, 2H), 3.24 - 3.02 (m, 2H), 2.16 - 1.72 (m, 4H), 1.43 (dd, 6H); LC-MS: m/z 368.36 (MH+).
Scheme 9, Cpd (IV)*a Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l- tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazol-3-yl]phenyl]methanone
Figure imgf000174_0001
In a proper vial palladium tritert-butylphosphine (4.42 mg, 0.010 mmol) and cesium fluoride (72.21 mg, 0.480 mmol) were placed and N2-vacuum cycles were performed 3 times. Then, a solution of trimethyl-[l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazol-3-yl]stannane (283.73 mg, 0.260 mmol) and [4-chloro-2-(l-propan-2-ylpyrazol-3-yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone
(85.0 mg, 0.220 mmol) in 1,4-Dioxane (3 mL) was added. The resulting suspension was degassed bubbling N2 for 5 min and then 5 cycles of N2-vacuum were performed. The mixture was stirred at 100 °C for 4h. After this time the mixture was cooled down to RT, diluted with EtOAc, filtered over a pad of celite washing with EtOAc and concentrated under high vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of
HCOOH to 65:35 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula (4,4-difluoro-l- piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazol-3- yl)phenyl)methanone.
Yield: 51 mg 1H NMR (400 MHz, DMSO) δ 7.98 (dd, 1H), 7.90 - 7.81 (m, 1H), 7.63 - 7.53 (m, 1H), 7.49 (dd, 1H),
7.12 (d, 1H), 6.54 (t, 1H), 5.46 - 5.37 (m, 1H), 4.52 (p, 1H), 4.11 - 3.95 (m, 1H), 3.89 - 3.76 (m, 1H),
3.75 - 3.49 (m, 2H), 3.26 - 3.21 (m, 1H), 3.21 - 3.11 (m, 1H), 2.43 - 2.35 (m, 1H), 2.18 - 1.80 (m, 5H),
1.74 - 1.47 (m, 5H), 1.47 - 1.33 (m, 6H); LC-MS: m/z 552.29 (MH+)
Scheme 9, Cpd (IV)*b Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5- isopropyl-l-tetrahydropyran-2-yl-pyrazol-3-yl)phenyl]methanone
Figure imgf000175_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound
(4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]phenyl]methanone wherein (5-isopropyl-l-tetrahydropyran-2-yl- pyrazol-3-yl)-trimethyl-stannane was used instead of trimethyl-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]stannane. The title compound was obtained in a yield of 59% (55 mg).
1H NMR (400 MHz, DMSO) δ 8.07 (d, 1H), 7.85 (d, 1H), 7.80 (d, 1H), 7.32 (d, 1H), 6.70 (s, 1H), 6.54
(d, 1H), 5.45 (d, 1H), 4.54 (p, 1H), 3.97 - 3.80 (m, 2H), 3.76 - 3.62 (m, 1H), 3.62 - 3.51 (m, 1H), 3.15 -
3.04 (m, 2H), 2.45 - 2.36 (m, 1H), 2.15 - 2.01 (m, 3H), 1.98 - 1.67 (m, 6H), 1.64 - 1.51 (m, 5H), 1.48 -
1.39 (m, 8H); LC-MS: m/z 526.33 (MH+).
Scheme 9, Cpd (IV)*c Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5- methyl-l-tetrahydropyran-2-yl-pyrazol-3-yl)phenyl]methanone
Figure imgf000175_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound
(4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]phenyl]methanone wherein trimethyl-(5-methyl-l-tetrahydropyran-
2-yl-pyrazol-3-yl)stannane was used instead of trimethyl-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]stannane. The title compound was obtained in a yield of 40% (24 mg).
1H NMR (400 MHz, DMSO) δ 8.05 (d, 1H), 7.84 (d, 1H), 7.77 (d, 1H), 7.32 (d, 1H), 6.65 (s, 1H), 6.51
(d, 1H), 5.43 (d, 1H), 4.53 (p, 1H), 3.92 (d, 2H), 3.69 (d, 1H), 3.57 (s, 1H), 3.23 - 3.04 (m, 2H), 2.40 -
2.31 (m, 3H), 2.12 - 2.01 (m, 2H), 1.97 - 1.51 (m, 7H), 1.46 - 1.42 (m, 7H); LC-MS: m/z 498.49 (MH+).
Scheme 9, Cpd (V)*a Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[5-
(trifluoromethyl)-lH-pyrazol-3-yl]phenyl]methanone
Figure imgf000176_0001
Example 84
To a solution of (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]phenyl]methanone (50.0 mg, 0.090 mmol) in Ethanol (2 mL) a 6 N aqueous solution of HCI (45.33 uL, 0.270 mmol) was added. The mixture was stirred at 60 °C for 2h.
After this time the reaction mixture was cooled down to RT, a s.s. of NaHCO3 was added and the mixture was extracted with EtOAc (3 times). The organic portions were combined, washed with brine, dried over Na2SO4 and concentrated under vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 55:45 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula (4,4-difluoro-l-piperidyl)-[2-(l- isopropylpyrazol-3-yl)-4-[5-(trifluoromethyl)-lH-pyrazol-3-yl]phenyl]methanone.
Yield: 20 mg
1H NMR (400 MHz, DMSO) δ 14.19 (s, 1H), 8.15 (d, 1H), 7.87 (d, 1H), 7.82 (dd, 1H), 7.42 (d, 1H), 7.33
(s, 1H), 6.62 (d, 1H), 4.52 (hept, 1H), 3.86 - 3.58 (m, 2H), 3.23 - 3.07 (m, 2H), 2.17 - 1.75 (m, 3H), 1.44
(dd, 7H); LC-MS: m/z 468.22 (MH+). Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5-isopropyl-lH-pyrazol-3- yl)phenyl]methanone
Figure imgf000177_0002
Example 85
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 84 (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[5-(trifluoromethyl)-lH- pyrazol-3-yl]phenyl]methanone wherein (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-
(5-isopropyl-l-tetrahydropyran-2-yl-pyrazol-3-yl)phenyl]methanone was used instead of (4,4- difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]phenyl]methanone. The title compound was obtained in a yield of
40% (15 mg).
1H NMR (400 MHz, DMSO) δ 12.67 (s, 1H), 8.07 (s, 1H), 7.84 (d, 1H), 7.76 (d, 1H), 7.30 (d, 1H), 6.56
(d, 2H), 4.52 (hept, 1H), 3.95 - 3.79 (m, 1H), 3.65 - 3.57 (m, 1H), 3.23 - 2.92 (m, 3H), 2.18 - 1.72 (m,
3H), 1.44 (dd, 7H), 1.28 (d, 6H); LC-MS: m/z 442.48 (MH+).
Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-lH-pyrazol-3- yl)phenyl]methanone
Figure imgf000177_0001
Example 86
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 84 (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[5-(trifluoromethyl)-lH- pyrazol-3-yl]phenyl]methanone wherein (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-
(5-methyl-l-tetrahydropyran-2-yl-pyrazol-3-yl)phenyl]methanone was used instead of (4,4- difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[l-tetrahydropyran-2-yl-5- (trifluoromethyl)pyrazol-3-yl]phenyl]methanone. The title compound was obtained in a yield of
66% (14 mg).
1H NMR (400 MHz, DMSO) δ 12.63 (s, 1H), 8.07 (s, 1H), 7.84 (d, 1H), 7.74 (d, 1H), 7.30 (d, 1H), 6.53
(d, 2H), 4.52 (hept, 1H), 3.92 - 3.75 (m, 1H), 3.73 - 3.52 (m, 1H), 3.24 - 3.06 (m, 2H), 2.29 (s, 3H), 2.15
- 1.71 (m, 3H), 1.44 (dd, 7H); LC-MS: m/z 414.47 (MH+).
Scheme 10
Figure imgf000178_0001
Step 1
Compound of formula II may be obtained via Stille coupling reaction between compound of formula
I (synthesized according to the Scheme 9) and trimethyl-[2-[(5-methyl-2-trimethylstannyl-imidazol- l-yl)methoxy]ethyl]silane, in the presence of a suitable catalyst, e.g. palladium tritertbutylphosphine, a suitable base, e.g. CsF, in a suitable solvent, e.g. 1,4-dioxane, typically at 100 °C.
The reaction takes about 5 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by removing the SEM protecting group under acidic conditions, e.g. HCI 6N solution, typically at 75 °C. The reaction takes about 16 hours to complete.
Scheme 10, Cpd (ll)*a Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[5- methyl-l-(2-trimethylsilylethoxymethyl)imidazol-2-yl]phenyl]methanone and (4,4-difluoro-l- piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[4-methyl-l-(2-trimethylsilylethoxymethyl)imidazol-2- yl]phenyl]methanone
Figure imgf000179_0001
In a proper vial palladium tritert-butylphosphine (3.21 mg, 0.010 mmol) and cesium fluoride (69.96 mg, 0.460 mmol) were placed and 3 cycles of N2-vacuum were performed. Then, a solution of trimethyl-[2-[(5-methyl-2-trimethylstannyl-imidazol-l-yl)methoxy]ethyl]silane/ trimethyl-[2-[(4- methyl-2-trimethylstannyl-imidazol-l-yl)methoxy]ethyl]silane (78.54 mg, 0.210 mmol) and [4- chloro-2-(l-propan-2-ylpyrazol-3-yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone (77.0 mg, 0.210 mmol) in 1,4-Dioxane (2.093 mL) was added. The resulting suspension was degassed bubbling N2 for 5 min and, then 5 cycles of N2-vacuum were performed. The mixture was stirred at 100 °C for
5h. After this time the mixture was cooled down to RT, diluted with EtOAc, filtered over a pad of celite washing with EtOAc and concentrated under high vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 45:55 of CH3CN/H2O + 0.1% of HCOOH) affording the products of formula (4,4-difluoro-l-piperidyl)-[2-(l- isopropylpyrazol-3-yl)-4-[5-methyl-l-(2-trimethylsilylethoxymethyl)imidazol-2- yl]phenyl]methanone and (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[4-methyl-l-(2- trimethylsilylethoxymethyl)imidazol-2-yl]phenyl]methanone as mixture of regioisomers. The mixture of regioisomers was used in the next step without further purification.
Yield: 18 mg
LC-MS: m/z 544.57 (MH+).
Scheme 10, Cpd (lll)*a Preparation: (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-(5- methyl-lH-imidazol-2-yl)phenyl]methanone
Figure imgf000180_0001
Example 87
To a solution of (4,4-difluoro-l-piperidyl)-[2-(l-isopropylpyrazol-3-yl)-4-[5-methyl-l-(2- trimethylsilylethoxymethyl)imidazol-2-yl]phenyl]methanone/ (4,4-difluoro-l-piperidyl)-[2-(l- isopropylpyrazol-3-yl)-4-[4-methyl-l-(2-trimethylsilylethoxymethyl)imidazol-2- yl]phenyl]methanone (mixture of regioisomers) (17.0 mg, 0.030 mmol) in Ethanol (1 mL) a 6 N aqueous solution of HCI (15.63 uL, 0.090 mmol) was added. The mixture was stirred for 16h at 75
°C. After this time the mixture was cooled down, a s.s. of NaHCO3 was added to the solution and the mixture was extracted with EtOAc (3X). The organic portions were combined, washed with brine, dried over Na2SO4 and concentrated under vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 30:70 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula (4,4-difluoro-l-piperidyl)-[2-(l- isopropylpyrazol-3-yl)-4-(5-methyl-lH-imidazol-2-yl)phenyl]methanone.
Yield: 8 mg
1H NMR (400 MHz, DMSO) δ 12.18 (d, 1H), 8.01 (d, 1H), 7.82 (d, 1H), 7.69 - 7.57 (m, 2H), 7.28 (d,
1H), 6.45 (d, 1H), 4.52 (hept, 1H), 3.98 - 3.80 (m, 1H), 3.68 - 3.48 (m, 1H), 3.23 - 3.07 (m, 2H), 2.45
(s, 3H), 2.16 - 1.71 (m, 3H), 1.44 (dd, 7H); LC-MS: m/z 414.28 (MH+).
Scheme 11
Figure imgf000180_0002
Step 1
Compound of formula II may be obtained via Suzuki coupling between compound of formula I
(synthesized according to the Scheme 3) and the desired boronic ester, such as 2-(cyclopenten-l- yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane, in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, a suitable base, e.g. Na2CO3, in a suitable mixture of solvents, e.g.
Toluene /Ethanol/ water, typically at 100 °C. The reaction takes from about 2 hours to 12 hours to complete.
Step 2
Compound of formula III may be obtained by reduction of double bond of compound of formula II under catalytic hydrogenation conditions, using for example Pd/C in a suitable solvent such as
Ethanol, typically at room temperature. The reaction takes about 2 hours to complete
Preparation: N-[3-(cyclopenten-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000181_0001
Example 88
In a vial were sequentially added palladium tetrakis triphenylphosphine (11.55 mg, 0.010 mmol), sodium carbonate (369.78 mg, 3.49 mmol) and 2-(cyclopenten-l-yl)-4,4,5,5-tetramethyl-
1,3,2-dioxaborolane (96.73 mg, 0.500 mmol). Solids were suspended in a mixture of Ethanol (1.7 mL), Toluene (1.7 mL) Water (0.500 mL) and the resulting suspension was degassed using shlenk line technique. N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide (200.0 mg, 0.500 mmol) was added under nitrogen flux.
The mixture was degassed again and heated to 100 °C for 2h. After this time the reaction mixture was cooled down to room temperature, diluted with water and extracted with EtOAc. The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum. The crude was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 40:60 of CH3CN/H2O + 0.1% of
HCOOH) affording the product of formula N-[3-(cyclopenten-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide.
Yield: 23 mg
1H NMR (400 MHz, DMSO-d6) δ 10.31 (s, 1H), 7.61 (d, 1H), 7.56 (dd, 1H), 7.28 (d, 1H), 5.88 (p, 1H),
4.28 (s, 1H), 3.73 (s, 1H), 3.47 (d, 3H), 3.14 (d, 3H), 2.88 (s, 1H), 2.68 - 2.52 (m, 1H), 2.46 (s, 2H), 1.97
- 1.84 (m, 2H), 1.77 (tt, 1H), 0.85 - 0.75 (m, 4H); LC-MS: m/z 389.2 (MH+). Preparation: N-[3-(cyclopenten-l-yl)-4-(4-methyl-2-phenylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000182_0002
Example 89
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 88 N-[3-(cyclopenten-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide wherein N-[3-bromo-4-(4-methyl-2-phenylpiperazine- l-carbonyl)phenyl]cyclopropanecarboxamide was used instead of N-[3-bromo-4-(l,l-dioxo-l,4- thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide. The title compound was obtained in a yield of 51 % (30 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.42 - 10.17 (m, 1H), 7.70 - 7.53 (m, 3H), 7.50 - 6.93 (m, 5H), 6.16 -
4.25 (m, 2H), 3.53 - 3.33 (m, 1H), 3.09 - 2.50 (m, 5H), 2.38 - 1.51 (m, 10H), 0.92 - 0.70 (m, 4H); LC-
MS: m/z 430.27 (MH+).
Preparation: N-[3-cyclopentyl-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000182_0001
Example 90
A mixture of N-[3-(cyclopenten-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide (23.0 mg, 0.060 mmol) and Pd-C 10% (6.3 mg, 0.010 mmol) in MeOH (5 mL) was stirred under H2 atmosphere (1 atm) at RT for 2 h. After this time the reaction was filtered and concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 60:40 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula N-[3-cyclopentyl-4-(l,l-dioxo- l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide. Yield: 7.6 mg
1H NMR (400 MHz, DMSO-d6) δ 10.24 (s, 1H), 7.63 (d, 1H), 7.48 (dd, 1H), 7.26 (d, 1H), 4.46 (d, 1H),
3.71 - 3.44 (m, 3H), 3.20 (d, 3H), 2.93 (p, 2H), 1.94 (dt, 2H), 1.75 (td, 3H), 1.61 (q, 2H), 1.47 (dq, 2H),
0.84 - 0.74 (m, 4H); LC-MS: m/z 391.5 (MH+).
Preparation: N-[3-cyclopentyl-4-(4-methyl-2-phenylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000183_0001
Example 91
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 90 N-[3-cyclopentyl-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide wherein N-[3-(cyclopenten-l-yl)-4-(4-methyl-2- phenylpiperazine-l-carbonyl)phenyl]cyclopropanecarboxamide was used instead of N-[3-
(cyclopenten-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide. The title compound was obtained in a yield of 54 % (16 mg).
NMR (400 MHz, DMSO-d6) δ 10.22 (s, 1H), 7.76 - 7.53 (m, 3H), 7.50 - 7.07 (m, 5H), 5.95 - 4.29 (m,
1H), 3.53 - 3.40 (m, 1H), 3.22 - 2.62 (m, 4H), 2.42 - 2.13 (m, 4H), 2.07 - 1.10 (m, 10H), 0.86 - 0.70 (m,
4H); LC-MS: m/z 432.29 (MH+)
Scheme 12
Figure imgf000183_0002
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Combi-Blocks) via Mitsunobu-Tsunoda reaction in presence of desired alcohol. The reaction is carried out in a suitable solvent, e.g. 1,4-Dioxane, under microwave irradiation, typically at 150° C.
The reaction takes about 30 minutes to complete. Alternatively compound of formula II may be obtained by N-alkylation of compound of formula I with a suitable electrophile reagent, e.g. a proper tosylate, in the presence of a suitable base, e.g. NaH, in a suitable solvent such as DMF, typically at 50 °C. The reaction takes about 12 hours to complete.
Step 2
Compound of formula III may be obtained via Miyaura Borylation between compound of formula II and B2Pin2, in presence of a suitable transition metal catalyst, e.g. Pd(dppf)Cl2, a suitable base, e.g. potassium acetate, in a suitable solvent, e.g. 1,4-Dioxane, typically at 100 °C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula III may be obtained from compound of formula IV (commercially available from Activate Scientific) via Mitsunobu-Tsunoda reaction in presence of desired alcohol in 1'4-
Dioxane as solvent, under microwave irradiation at 150° C. The reaction takes about 30 minutes to complete.
Scheme 12, Cpd (ll)*a Preparation: l-cyclobutyl-3-iodopyrazole
Figure imgf000184_0001
A mixture of 3-iodo-lH-pyrazole (1.0 g, 5.16 mmol), cyclobutanol (371.76 mg, 5.16 mmol), 2- tributylphosphoranylideneacetonitrile (2.7 mL, 10.31 mmol) in 1,4-Dioxane (17.78 mL) was stirred under microwave irradiation at 150 °C for 30 minutes. The mixture was diluted with water (20 mL) and extracted with EtOAc. The organic phase was washed with brine, dried over Na2SO4, filtered and then concentrated under high vacuum. The crude was purified FC on silica gel (eluting from
100% of cHex to cHex/EtOAc 90:10) affording the product of formula l-cyclobutyl-3-iodopyrazole. Yield: 295 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 7.27 (d, 1H), 6.40 (d, 1H), 4.82 - 4.70 (m, 1H), 2.61 - 2.39 (m,
4H), 1.95 - 1.79 (m, 2H); LC-MS: m/z 249.06 (MH+).
Scheme 12, Cpd (ll)*b Preparation: l-(l-cyclopropylethyl)-3-iodopyrazole
Figure imgf000185_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 1- cyclobutyl-3-iodopyrazole wherein 1-cyclopropylethanol was used instead of cyclobutanol. The title compound was obtained in in a yield of 44 % (877 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.56 (dd, 1H), 6.43 (t, 1H), 3.81 (dq, 1H), 1.46 (dd, 3H), 1.40 -
1.29 (m, 1H), 0.62 - 0.22 (m, 4H); LC-MS: m/z 263.1 (MH+).
Scheme 12, Cpd (ll)*c Preparation: l-tert-butyl-3-iodopyrazole
Figure imgf000185_0002
To a solution of 3-iodo-lH-pyrazole (1.0 g, 5.16 mmol) in 2-methyl-2-propanol (5.08 mL, 53.1 mmol), H2SO4 95-97% (0.35 mL, 6.19 mmol) was added. The mixture was stirred at 100 °C overnight. After this time the mixture was diluted with a saturated solution of NaHCO3 until pH = 7 and extracted with EtOAc. Organic phase was washed with brine, dried over Na2SO4 and concentrated to give a crude that was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc 80:20) to afford the product of formula l-tert-butyl-3-iodopyrazole.
Yield: 713 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 7.73 (d, 1H), 6.43 (d, 1H), 1.49 (s, 9H); LC-MS: m/z
251.00 (MH+). Scheme 12, Cpd (ll)*d Preparation: 3-iodo-l-(oxetan-3-yl)pyrazole
Figure imgf000186_0001
To a solution of 3-iodo-lH-pyrazole (0.51 g, 2.63 mmol) in DMF (10 mL), sodium hydride (0.42 g,
10.52 mmol) was added at 0 °C and the reaction was stirred at the same temperature for 10 min.
After this time oxetan-3-yl 4-methylbenzenesulfonate (0.9 g, 3.94 mmol) was added. The reaction was allowed to reach RT and, then, stirred at 50 °C overnight. After this time the mixture was diluted with water and extracted with EtOAt. The organic phase was washed with Brine, dried over Na2SO4 and concentrated under reduced pressure. The crude was purified by FC on NH column (eluting from 100% of cHex to cHex/EtOAc 90:10) to afford the product of formula 3-iodo-l-(oxetan-3-yl) pyrazole.
Yield: 445 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 7.80 (d, 1H), 6.52 (d, 1H), 5.57 (tt, 1H), 4.91 - 4.81 (m, 4 H); LC-
MS: m/z 251.12 (MH+).
Preparation: oxetan-3-yl 4-methylbenzenesulfonate
Figure imgf000186_0002
To a solution of 3-oxetanol (1.0 g, 13.5 mmol) in DCM (20mL), Pyridine (2.17 mL, 27 mmol) was added followed by DMAP (0.33 g, 2.7 mmol) and 4-methylbenzenesulfonyl chloride (2.83 g, 14.85 mmol). The reaction was allowed to stir overnight at room temperature. After this time the mixture was diluted with a IN aqueous solution of HCI and extracted with DCM. Organic phase was washed with a s.s. of NaHCO3. The organic phase was dried using a phase separator and concentrated to afford a crude that was purified by FC on silica gel (eluting from 100% of cHex 100% to cHex/EtOAc
80:20) to afford the product of formula oxetan-3-yl 4-methylbenzenesulfonate.
Yield: 1.96 g.
1H NMR (400 MHz, DMSO-d6) δ ppm 7.80 (d, 2H), 7.49 (d, 2H), 5.39 - 5.31 (m, 1H), 4.71 - 4.63 (m,
2H), 4.49 - 4.37 (m, 2H), 2.42 (s, 3H); LC-MS: m/z 229.07 (MH+).
Scheme 12, Cpd (lll)*a Preparation: l-cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazole
Figure imgf000187_0001
A mixture of l-cyclobutyl-3-iodopyrazole (290.0 mg, 1.17 mmol), 4,4,5, 5-tetramethyl-2-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (356.25 mg, 1.4 mmol), potassium acetate (401.56 mg, 4.09 mmol) and [l,l'-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (382.82 mg, 0.470 mmol) in 1,4-Dioxane (5 mL) was degassed with N2 for 5 min followed by 3 cycles of vacuum/N2. The reaction was stirred at 100 °C overnight. The day after the mixture was cooled down to room temperature and diluted with water. The mixture was extracted with EtOAc 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula 1- cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole (see Scheme 12, Cpd (lll)*a).
The product was used as such in the next step.
Yield: 314.6 mg
LC-MS: m/z 167.06 (MH+) mass of boronic acid.
Scheme 12, Cpd (lll)*b Preparation: l-(l-cyclopropylethyl)-3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazole
Figure imgf000188_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 12, Cpd (lll)*a l-cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole wherein l-(l-cyclopropylethyl)-3-iodopyrazole was used instead of l-cyclobutyl-3-iodopyrazole.
The title compound was obtained in a yield of 76 % (459 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.79 (d, 1H), 6.58 (d, 1H), 4.20 - 4.09 (m, 1H), 1.55 - 1.43 (m,
3H), 1.25 - 1.20 (m, 1H), 1.07 (s, 12H), 0.63 - 0.17 (m, 4H); LC-MS: m/z 181.1 (MH+) mass of boronic acid.
Scheme 12, Cpd (lll)*c Preparation: l-tert-butyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazole
Figure imgf000188_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 12, Cpd (lll)*a l-cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole wherein l-tert-butyl-3-iodopyrazole was used instead of l-cyclobutyl-3-iodopyrazole. The title compound was obtained in a quantitative yield (472 mg).
LC-MS: m/z 167.06 (MH+) mass of boronic acid.
Scheme 12, Cpd (lll)*d Preparation: l-(oxetan-3-yl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazole
Figure imgf000189_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 12, Cpd (lll)*a l-cyclobutyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole wherein 3-iodo-l-(oxetan-3-yl)pyrazole was used instead of l-cyclobutyl-3-iodopyrazole. The title compound was obtained in a quantitative yield (228 mg).
LC-MS: m/z 247.18 (MH+)
Scheme 12, Cpd (lll)*e Preparation: l-(2-methylpropyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-
2-yl)pyrazole
Figure imgf000189_0002
A mixture of 2-methyl-l-propanol (0.57 mL, 6.18 mmol), 3-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (1.2 g, 6.18 mmol) and 2-tributylphosphoranylideneacetonitrile
(3.24 mL, 12.37 mmol) in 1,4-Dioxane (12 mL) was stirred at 150 °C for 30 min under microwave irradiation. The reaction mixture was cooled down to room temperature and diluted with water and extracted with EtOAc (3x). The organic layers were collected together and washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum affording the product of formula l-(2-methylpropyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole (see Scheme 12, Cpd
(lll)*e), which was used in the next step without further purifications.
Yield: 1 g. 1H NMR (400 MHz, MeOD) δ ppm 7.48 (d, 1H), 6.68 (d, 1H), 4.21 (d, 2H), 2.13 (dp, 1H), 1.35 (s, 12H),
0.86 (d, 6H); LC-MS: m/z 169.0 (MH+) mass of boronic acid.
Scheme 12, Cpd (III) *f Preparation: l-ethyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole
Figure imgf000190_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 12, Cpd (lll)*e l-(2-methylpropyl)-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole wherein ethanol was used instead of 2-methyl-l-propanol. The title compound was obtained in a quantitative yield (2.99 g).
LC-MS: m/z 141.01 (MH+) mass of boronic acid.
Scheme 13
Figure imgf000190_0002
Step 1
Compound of formula II may be obtained by N-protection of compound of formula I with 3,4- dihydro-2H-pyran in the presence of a catalytic amount of 4-toluensulfonic acid, in a suitable solvent, e.g. DCM, typically at RT. The reaction takes about 12 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by reaction with trimethyl(trimethylstannyl)stannane in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, a suitable solvent, e.g. toluene, typically at 100 °C. The reaction takes from about 1 hour to 20 hours to complete. Scheme 13, Cpd (ll)*a Preparation: 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole
Figure imgf000191_0001
To a stirred solution of 3-bromo-5-(trifluoromethyl)-lH-pyrazole (200.0 mg, 0.930 mmol) in DCM
(9.304 mL), 3,4-dihydro-2H-pyran (126.5 uL, 1.4 mmol) and 4-toluensulfonic acid (16.02 mg, 0.090 mmol) were added. The mixture was stirred overnight at RT. After this time, the mixture was washed with a s.s. of NaHCO3 and the aqueous phase was counterextracted with DCM. The organic phases were combined, washed with brine, dried over a phase separator and concentrated under vacuum. The crude was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc 85:15) affording the product of formula 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole (see
Scheme 13, Cpd (ll)*a).
Yield: 218 mg
1H NMR (400 MHz, DMSO-d6) δ 7.14 (d, 1H), 5.59 (dd, 1H), 3.98 - 3.87 (m, 1H), 3.72 - 3.58 (m, 1H),
2.30 - 2.15 (m, 1H), 2.04 - 1.86 (m, 2H), 1.81 - 1.64 (m, 1H), 1.60 - 1.50 (m, 2H).
Scheme 13, Cpd (ll)*b Preparation: 3-iodo-l-(oxan-2-yl)-5-propan-2-ylpyrazole
Figure imgf000191_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 13, Cpd (ll)*a 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole wherein 3- iodo-5-propan-2-yl-lH-pyrazole was used instead of 3-bromo-5-(trifluoromethyl)-lH-pyrazole. The title compound was obtained in yield of 94% (320 mg).
LC-MS: m/z 321.07
Scheme 13, Cpd (ll)*c Preparation: 3-iodo-5-methyl-l-(oxan-2-yl)pyrazole
Figure imgf000192_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 13, Cpd (ll)*a 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole wherein 3- iodo-5-propan-2-yl-lH-pyrazole was used instead of 3-bromo-5-(trifluoromethyl)-lH-pyrazole. The title compound was obtained in yield of 79% (560 mg).
LC-MS: m/z 293.14 (MH+).
Scheme 13, Cpd (lll)*a Preparation: trimethyl-[l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazol-
3-yl]stannane
Figure imgf000192_0001
In a proper vial trimethyl(trimethylstannyl)stannane (178.87 uL, 0.860 mmol), 3-bromo-l- tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole (215.0 mg, 0.720 mmol, see Scheme 13, Cpd
(ll)*a), palladium tetrakis triphenylphosphine (16.61 mg, 0.010 mmol) were placed and suspended in Toluene (5 mL). The mixture was degassed bubbling N2 for 5 min and then 5 cycles of N2-vacuum were performed. The mixture was stirred at 100 °C for 20h. After this time the reaction was cooled down to RT, filtered and concentrated under vacuum. The residue was dissolved in EtOAc and washed with water. The aqueous phase was counterextracted with EtOAc (X2). The combined organic portions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula trimethyl-[l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazol-3-yl]stannane, which was used in the next step without further purification.
Yield: 270 mg
LC-MS: m/z 385.11 (MH+). Scheme 13, Cpd (lll)*b Preparation: (5-isopropyl-l-tetrahydropyran-2-yl-pyrazol-3-yl)-trimethyl- stannane
Figure imgf000193_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 13, Cpd (ll)*a 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole wherein 3- iodo-l-(oxan-2-yl)-5-propan-2-ylpyrazole was used instead of the 3-bromo-l-tetrahydropyran-2-yl-
5-(trifluoromethyl)pyrazole. The title compound was obtained in yield of 83% (250 mg).
LC-MS: m/z 359.18 (MH+).
Scheme 13, Cpd (lll)*c Preparation: trimethyl-(5-methyl-l-tetrahydropyran-2-yl-pyrazol-3- yl)stannane
Figure imgf000193_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 13, Cpd (ll)*a 3-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)pyrazole wherein 3- iodo-5-methyl-l-(oxan-2-yl)pyrazole was used instead of the 3-bromo-l-tetrahydropyran-2-yl-5-
(trifluoromethyl)pyrazole. The title compound was obtained in yield of 88% (150 mg).
LC-MS: m/z 331.20 (MH+).
Scheme 14
Figure imgf000193_0003
Step 1
Compound of formula II may be obtained via N-protection of compound of formula I with SEM-CI, in the presence of a suitable base, e.g. K2CO3, in a suitable solvent, e.g. acetone, typically at RT. The reaction takes about 2 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by reaction with trimethyl(trimethylstannyl)stannane in the presence of a suitable catalyst, e.g. palladium tetrakis triphenylphosphine, a suitable solvent, e.g. toluene, typically at 100 °C. The reaction takes from about 3 hours to complete.
Scheme 14, Cpd (II) Preparation: 2-[(2-iodo-5-methyl-imidazol-l-yl)methoxy]ethyl-trimethyl-silane and 2-[(2-iodo-4-methyl-imidazol-l-yl)methoxy]ethyl-trimethyl-silane
Figure imgf000194_0001
A mixture of 2-iodo-5-methyl-lH-imidazole (200.0 mg, 0.960 mmol) and potassium carbonate
(398.68 mg, 2.88 mmol) in Acetone (8.013 mL) was treated by dropwise addition of 2-
(chloromethoxy)ethyl-trimethylsilane (204.21 uL, 1.15 mmol) and the resulting mixture was stirred for 2h. After this time, the mixture was filtered and concentrated under vacuum. The crude was purified by FC on silica gel (eluting from cHex/EtOAc 90:10 to cHex/EtOAc 30:70) affording the products of formula 2-[(2-iodo-5-methyl-imidazol-l-yl)methoxy]ethyl-trimethyl-silane and 2-[(2- iodo-4-methyl-imidazol-l-yl)methoxy]ethyl-trimethyl-silane as mixture as regioisomers. The mixture was used in the next step without further purification.
Yield: 100 mg
LC-MS: m/z 339.25 (MH+).
Scheme 14, Cpd (lll) Preparation: trimethyl-[2-[(5-methyl-2-trimethylstannyl-imidazol-l- yl)methoxy]ethyl]silane and trimethyl-[2-[(4-methyl-2-trimethylstannyl-imidazol-l- yl)methoxy]ethyl]silane
Figure imgf000195_0001
A mixture of trimethyl(trimethylstannyl)stannane (40.46 uL, 0.200 mmol), 2-[(2-iodo-5-methyl- imidazol-l-yl)methoxy]ethyl-trimethyl-silane (55.0 mg, 0.160 mmol) and palladium tetrakis triphenylphosphine (3.76 mg, 0 mmol) in Toluene (1.7 mL) was degassed bubbling N2 for 15 min.
Then 5 cycles of N2-vacuum were performed and the mixture was stirred at 100 °C for 3h. The mixture was concentrated under vacuum, the residue was dissolved in EtOAc and washed with water. The aqueous phase was extracted with EtOAc (X2). The combined organic portions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the products of formula trimethyl-[2-[(5-methyl-2-trimethylstannyl-imidazol-l-yl)methoxy]ethyl]silane and trimethyl-[2-[(4-methyl-2-trimethylstannyl-imidazol-l-yl)methoxy]ethyl]silane as mixture of regioisomers, The mixture was used in the next step without further purification.
Yield: 65 mg
LC-MS: m/z 377.2 (MH+)
Scheme 15
Figure imgf000195_0002
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Combi-Blocks) by hydroxycarbonylation using oxalic acid and acetic anhydride as the in situ carbon monoxide generator. The reaction is carried out in the presence of a suitable organic base, e.g. DIPEA, a suitable catalyst, e.g. Palladium diacetate, with a suitable phosphine ligand, e.g. Xantphos; in a suitable solvent such as DMF, typically at 100 °C. The reaction takes about 12 hours to complete.
Step 2
Compound of formula III may be obtained via acyl chloride formation of compound of formula II by treatment with a suitable chlorinating agent, such as SOCl2, typically at 90 °C, followed by coupling with the proper amine in the presence of a suitable organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes from about
1 hour to about 12 hours to complete.
Step 3
Compound of formula IV may be obtained from compound of formula III by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes from about 12 hours to about 24 hours to complete.
Step 4
Compound of formula V may be obtained by nitro reduction of compound of formula IV under catalytic hydrogenation conditions, using a suitable catalyst, such as Pd/C, in a suitable solvent such as methanol, typically at room temperature. The reaction takes from about 4 hours to about 12 hours to complete.
Step 5
Compound of formula VI may be obtained by coupling reaction between compound of formula V and a proper commercially available acyl chloride, e.g. cyclopropanecarbonyl chloride, in presence of a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes from about 1 hour to about 12 hours to complete.
Scheme 15, Cpd (ll)*a Preparation: 2-fluoro-5-methyl-4-nitrobenzoic acid
Figure imgf000196_0001
In a 40 mL vial equipped with magnetic bar l-bromo-2-fluoro-5-methyl-4-nitrobenzene (500.0 mg,
2.14 mmol), oxalic acid (288.53 mg, 3.2 mmol), [Pd(AcO)2]3 (4.8 mg, 0.020 mmol) and (5- diphenylphosphino-9,9-dimethyl-4-xanthenyl)-diphenylphosphine (12.36 mg, 0.020 mmol) were introduced. A solution of Acetic acid acetyl ester (0.3 mL, 3.2 mmol) and N,N-Diisopropylethylamine
(0.56 mL, 3.2 mmol) in DMF (11 mL) was added and the mixture was bubbled with N2 for 10 min and then 3 cycles of N2-vacuum were performed. The mixture was stirred at 100 °C overnight. After this time a s.s of NaHCO3 was added and the aqueous phase was washed with DCM. The aqueous phase was then acidified with concentrated HCI until pH = 1 and extracted with EtOAc. The organic fractions were combined, washed with brine, dried over Na2SO4 and then concentrated under vacuum to afford the product of formula 2-fluoro-5-methyl-4-nitrobenzoic acid (see Scheme 15,
Cpd (ll)*a), which was used in the next step without further purification.
Yield: 410 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.32 - 7.83 (m, 2H), 2.51 (s, 3H); LC-MS: m/z 198.18 (MH+).
Scheme 15, Cpd (ll)*b Preparation: 2-fluoro-3-methyl-4-nitrobenzoic acid
Figure imgf000197_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Scheme 15, Cpd (ll)*a 2-fluoro-5-methyl-4-nitrobenzoic acid wherein l-bromo-2-fluoro-3-methyl-
4-nitrobenzene was used instead of l-bromo-2-fluoro-5-methyl-4-nitrobenzene. The title compound was obtained in a yield of 68% (289 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 13.81 (s, 1H), 7.91 - 7.83 (m, 2H), 2.39 (s, 3H); LC-MS: m/z
198.17 (MH+).
Scheme 15, Cpd (lll)*a Preparation: (2-fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000197_0001
A mixture of 2-fluoro-5-methyl-4-nitrobenzoic acid (205.0 mg, 1.03 mmol, Scheme 15, Cpd (ll)*a) and thionyl dichloride (1.5 mL, 20.59 mmol) was heated to reflux for 2 hours. After this time the reaction was cooled down to RT and then concentrated under vacuum. The mixture was stripped three times with cyclohexane in order to remove the excess of thionyl dichloride. The residue was suspended in dry DCM (4.1 mL) and triethylamine (0.43 mL, 3.09 mmol) was added followed by 1- methyl-3-phenylpiperazine (181.45 mg, 1.03 mmol). The reaction mixture was stirred overnight at
RT. The day after the reaction was diluted with DCM and washed with a s.s of NaHCO3 3 times. The aqueous phases were combined and counterextracted with DCM. The combined organic phases were washed with brine, dried over a phase separator and concentrated under vacuum affording the product of formula (2-fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone. The product was used in the next step without further purification.
Yield: 297 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.07 (d, 1H), 7.68 (s, 1H), 7.55 - 7.03 (m, 5H), 5.90 - 5.33 (m,
1H), 4.92 - 4.30 (m, 1H), 4.00 - 3.43 (m, 2H), 3.26 - 2.71 (m, 3H), 2.53 (s, 3H), 2.29 - 2.17 (m, 3H); LC-
MS: m/z 358.36 (MH+).
Scheme 15. Cpd (lll*b Preparation: (l,l-dioxo-l,4-thiazinan-4-yl)-(2-fluoro-5-methyl-4- nitrophenyl)methanone
Figure imgf000198_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound (2- fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein 1,4- thiazinane 1,1-dioxide hydrochloride was used instead of l-methyl-3-phenylpiperazine. The title compound was obtained in a yield of 81% (297 mg).
NMR (400 MHz, DMSO-d6) δ 8.08 (d, 1H), 7.78 (d, 1H), 4.58 - 4.16 (m, 1H), 3.85 - 3.58 (m, 3H),
3.19 - 2.92 (m, 4H), 2.51 (s, 3H). ; LC-MS: m/z 317.10 (MH+).
Scheme 15, Cpd (Ill)*c Preparation: (2-fluoro-3-methyl-4-nitrophenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000199_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound (2- fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein 2-fluoro-3- methyl-4-nitrobenzoic acid was used instead of 2-fluoro-5-methyl-4-nitrobenzoic acid (see Scheme
15, Cpd (ll)*a). The title compound was obtained in a quantitative yield (272 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.99 - 7.78 (m, 1H), 7.66 - 7.15 (m, 6H), 5.79 (s, 1H), 4.73 -4.34
(m, 1H), 3.62 (dd, 1H), 3.26 - 2.69 (m, 4H), 2.42 (s, 2H), 2.25 - 2.13 (m, 3H), 2.01 - 1.88 (m, 1H); LC-
MS: m/z 358.36 (MH+).
Scheme 15. Cpd (lll)*d Preparation: (l,l-dioxo-l,4-thiazinan-4-yl)-(2-fluoro-3-methyl-4- nitrophenyl)methanone
Figure imgf000199_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound (2- fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein 1,4- thiazinane 1,1-dioxide hydrochloride was used instead of l-methyl-3-phenylpiperazine and
2-fluoro-3-methyl-4-nitrobenzoic acid was used instead of 2-fluoro-5-methyl-4-nitrobenzoic acid
(see Scheme 15, Cpd (ll)*a). The title compound was obtained in a yield of 84% (193 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.95 (dd, 1H), 7.80 - 7.54 (m, 1H), 4.34 (d, 2H), 3.65 (s, 4H),
2.98 (d, 2H), 2.41 (d, 3H); LC-MS: m/z 317.09 (MH+).
Scheme 15, Cpd (IV)*a Preparation: (5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000200_0001
To a solution of (2-fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone
(290.0 mg, 0.810 mmol) in DMSO (4.0 mL), pyrrolidine (0.2 mL, 2.43 mmol) was added and the reaction mixture was stirred overnight at 120 °C. After this time the reaction mixture was cooled down to RT, diluted with H2O and extracted three times with EtOAc. The organic portions were combined, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula (5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone. The crude was used in the next step without further purification.
Yield: 351 mg
LC-MS: m/z 409.27 (MH+).
Scheme 15, Cpd (IV)*b Preparation: (l,l-dioxo-l,4-thiazinan-4-yl)-(5-methyl-4-nitro-2-pyrrolidin-l- ylphenyl)methanone
Figure imgf000200_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound (5- methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein
(l,l-dioxo-l,4-thiazinan-4-yl)-(2-fluoro-5-methyl-4-nitrophenyl)methanone was used instead of (2- fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone. The title compound was obtained in a yield of 92% (247 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.38 (d, 1H), 7.18 (s, 1H), 4.52 (d, 1H), 3.77 (d, 1H), 3.69 - 3.49
(m, 2H), 3.40 - 3.35 (m, 2H), 3.27 - 3.01 (m, 6H), 2.37 (s, 3H), 1.90 (d, 4H); LC-MS: m/z 368.13 (MH+).
Scheme 15, Cpd (IV)*c Preparation: (l,l-dioxo-l,4-thiazinan-4-yl)-(3-methyl-4-nitro-2-pyrrolidin-l- ylphenyl)methanone
Figure imgf000201_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound (5- methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein
(l,l-dioxo-l,4-thiazinan-4-yl)-(2-fluoro-3-methyl-4-nitrophenyl)methanone was used instead of (2- fluoro-5-methyl-4-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone. The title compound was obtained in a yield of 37% (82 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.63 (d, 1H), 7.47 (d, 1H), 4.58 (d, 1H), 3.73 (d, 1H), 3.63 - 3.33
(m, 4H), 3.26 - 2.95 (m, 6H), 2.28 (s, 3H), 1.93 - 1.77 (m, 4H); LC-MS: m/z 368.2 (MH+).
Scheme 15, Cpd (V)*a Preparation: (4-amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000201_0002
A mixture of (5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone (350.0 mg, 0.860 mmol) and palladium (182.36 mg, 0.090 mmol) was stirred under H2 atmosphere (1 atm) at RT for 4h. After this time the reaction was filtered and concentrated under vacuum affording the product of formula (4-amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone. The product was used in the next step without further purification.
Yield: 292 mg
LC-MS: m/z 379.3 (MH+).
Scheme 15, Cpd (V)*b Preparation: (4-amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(l,l-dioxo-l,4- thiazinan-4-yl)methanone
Figure imgf000202_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound (4- amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein
(l,l-dioxo-l,4-thiazinan-4-yl)-(5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)methanone was used instead of (5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone. The title compound was obtained in a yield of 96% (215 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 6.76 (d, 1H), 6.04 (s, 1H), 4.95 (s, 2H), 4.22 (s, 1H), 3.96 - 3.45
(m, 3H), 3.27 - 2.92 (m, 8H), 1.96 (s, 3H), 1.89 - 1.77 (m, 4H); LC-MS: m/z 338.17 (MH+).
Scheme 15, Cpd (V)*c Preparation: (4-amino-3-methyl-2-pyrrolidin-l-ylphenyl)-(l,l-dioxo-l,4- thiazinan-4-yl)methanone
Figure imgf000202_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound (4- amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone wherein
(l,l-dioxo-l,4-thiazinan-4-yl)-(3-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)methanone was used instead of (5-methyl-4-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone. The title compound was obtained in a quantitative yield (95 mg).
LC-MS: m/z 338.2 (MH+).
Scheme 15, Cpd (Vl)*a Preparation: N-[2-methyl-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000203_0001
Example 92
To a mixture of N,N-Diisopropylethylamine (0.27 mL, 1.54 mmol) and (4-amino-5-methyl-2- pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone (292.0 mg, 0.770 mmol) in DCM (20 mL) at 0 °C, cyclopropanecarbonyl chloride (84.0 uL, 0.930 mmol) was added dropwise. The mixture was stirred overnight at RT. Then MeOH was added and concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted three times with AcOEt. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula N-[2- methyl-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide.
Yield: 138 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.50 - 9.23 (m, 1H), 7.82 - 7.16 (m, 5H), 7.09 - 6.68 (m, 2H),
5.86 - 4.34 (m, 1H), 3.63 - 3.35 (m, 1H), 3.28 - 2.57 (m, 6H), 2.42 - 2.05 (m, 7H), 1.93 (d, 5H), 1.63
(dt, 2H), 0.85 - 0.67 (m, 4H); LC-MS: m/z 447.3 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-2-methyl-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000203_0002
Example 93
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 92 N-[2-methyl-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein (4-amino-5-methyl-2-pyrrolidin-l-ylphenyl)-(l,l- dioxo-l,4-thiazinan-4-yl)methanone was used instead of (4-amino-5-methyl-2-pyrrolidin-l- ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone. The title compound was obtained in a yield of 86% (224 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 9.37 (s, 1H), 7.01 (d, 2H), 4.38 (d, 1H), 3.89 - 3.45 (m, 3H), 3.25
- 2.98 (m, 8H), 2.12 (s, 3H), 1.97 - 1.77 (m, 5H), 0.84 - 0.71 (m, 4H); LC-MS: m/z 406.2 (MH+).
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-2-methyl-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000204_0001
Example 94
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 92 N-[2-methyl-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein (4-amino-3-methyl-2-pyrrolidin-l-ylphenyl)-(l,l- dioxo-l,4-thiazinan-4-yl)methanone was used instead of (4-amino-5-methyl-2-pyrrolidin-l- ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone. The title compound was obtained in a yield of 19% (22 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 9.55 (s, 1H), 7.21 (d, 1H), 7.13 (d, 1H), 4.53 (d, 1H), 3.76 (d,
1H), 3.60 - 3.46 (m, 1H), 3.42 - 3.33 (m, 2H), 3.28 - 3.13 (m, 2H), 3.11 - 2.91 (m, 5H), 2.10 (s, 3H),
1.94 - 1.71 (m, 5H), 0.79 (d, 4H); LC-MS: m/z 406.2 (MH+).
Scheme 16
Figure imgf000205_0001
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Fluorochem) by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. CH3CN, typically at 80 °C. The reaction takes about 1 hour to complete.
Step 2
Compound of formula III may be obtained by nitro reduction of compound of formula II using Zn dust in a suitable solvent, e.g. AcOH, typically at room temperature. The reaction takes about 4 hours to complete.
Step 3
Compound of formula IV may be obtained by coupling reaction between compound of formula III and a proper commercially available acyl chloride, e.g. cyclopropanecarbonyl chloride, in presence of a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 12 hours to complete.
Step 4
Compound of formula V may be obtained by chlorination of compound of formula IV using N-
Chlorosuccinimide in a suitable solvent such as 2-propanol, typically at 60 °C. The reaction takes about 2 hours to complete. Step 5
Compound of formula VI may be obtained from compound of formula V by hydrolysis, e.g. using
LiOH, in a suitable mixture of solvents such as MeOH/THF/H2O, typically at 50 °C. The reaction takes from about 3 hours to about 24 hours to complete.
Step 6
Compound of formula VII may be obtained from compound of formula VI by coupling with a proper amine in the presence of coupling agent, e.g. HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes from about 1 hour to about 30 hours to complete.
Scheme 16, Cpd (II) Preparation: methyl 4-nitro-2-pyrrolidin-l-ylbenzoate
Figure imgf000206_0001
To a solution of 2-fluoro-4-nitrobenzoate (5.0 g, 25.11 mmol) in CH3CN (50 mL) , pyrrolidine (5.36 g,
75.32 mmol) was added and the reaction mixture was stirred at 80 °C for 1 h. After this time the mixture was cooled down to RT and then concentrated under vacuum. The residue was suspended in water and filtered. The solid was dried under vacuum affording the product of formula methyl 4- nitro-2-pyrrolidin-l-ylbenzoate, which was used in the next step without further purification.
Yield: 6.0 g
1H NMR (400 MHz, DMSO-d6) δ ppm 7.64 (d, 1H), 7.50 (d, 1H), 7.44 (dd, 1H), 3.86 (s, 3 ), 3.19 - 3.27
(m, 4H), 1.88 - 1.99 (m, 4H); LC-MS: m/z 251.10 (MH+).
Scheme 16, Cpd (III) Preparation: methyl 4-amino-2-pyrrolidin-l-ylbenzoate
Figure imgf000206_0002
To a solution of methyl 4-nitro-2-pyrrolidin-l-ylbenzoate (6.0 g, 22.06 mmol) in Acetic acid (150 mL), zinc (14.4 g, 220.58 mmol) was added. The reaction was stirred at RT for 4 h. After this time the reaction was filtered washing with EtOAc and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted three times with EtOAc. The organic portions were combined, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula methyl 4-amino-2-pyrrolidin-l-ylbenzoate, which was used in the next step without further purifications.
Yield: 5.75 g
1H NMR (400 MHz, DMSO-d6) δ ppm 7.28 (d, 1H), 5.98 (d, 1H), 5.94 (dd, 1H), 5.46 (s, 2H), 3.67 (s,
3H), 3.18 - 2.94 (m, 4H), 1.91 - 1.71 (m, 4H); LC-MS: m/z 221.99 (MH+).
Scheme 16, Cpd (IV) Preparation: methyl 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoate
Figure imgf000207_0001
To a stirred solution of methyl 4-amino-2-pyrrolidin-l-ylbenzoate (5.75 g, 26.1 mmol) and N,N-
Diisopropylethylamine (9.09 mL, 52.21 mmol) in DCM (145.57 mL) at 0 °C, cyclopropanecarbonyl chloride (3.08 mL, 33.94 mmol) was added dropwise and the reaction mixture was stirred at RT overnight. Reaction was diluted with DCM and washed twice with a s.s. of NaHCO3. The organic portion was washed with water and then with brine, dried over a phase separator and the solvent was evaporated under vacuum. The crude was purified by FC on silica gel (eluting from cHex/EtOAc 9:1 to cHex/EtOAc 50:50) to afford the product of formula methyl 4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate.
Yield: 5.57 g
1H NMR (400 MHz, DMSO-d6) δ ppm 10.21 (s, 1H), 7.41 (d, 1H), 7.22 (d, 1H), 6.92 (dd, 1H), 3.75 (s,
3H), 3.16 - 3.02 (m, 4H), 1.93 - 1.73 (m, 5H), 0.84 - 0.76 (m, 4H); LC-MS: m/z 288.74 (MH+).
Scheme 16, Cpd (V) Preparation: methyl 5-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoate and methyl 3-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate
Figure imgf000208_0001
1 -Chloropyrrolidine-2, 5-dione (272.3 mg, 2.04 mmol) was added one-pot to a stirred solution of methyl 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate (490.0 mg, 1.7 mmol) in 2-
Propanol (15 mL). The reaction was stirred at 60 °C for 2 hours. After this time, the mixture was cooled down to RT, water was added and the mixture was concentrated under high vacuum. The residue was dissolved in EtOAc and washed with a s.s of NaHCO3. The aqueous phase was extracted twice with EtoAc. Organic portions were combined, washed with brine, dried over Na2SO4 and concentrated under vacuum. The crude was purified by FC on silica gel (eluting from Chex 100% to
Chex/EtOAc 80:20) affording the products of formula methyl 5-chloro-4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate and methyl 3-chloro-4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate .
Yield of methyl 5-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate : 175 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.60 (s, 1H), 7.51 (s, 1H), 7.46 (s, 1H), 3.79 (s, 3H), 3.18 - 2.95
(m, 4H), 2.08 (dd, 1H), 1.87 (q, 4H), 0.94 - 0.66 (m, 4H); LC-MS: m/z 323.14 (MH+).
Yield of methyl 3-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate: 80 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.75 (s, 1H), 7.64 (d, 1H), 7.44 (d, 1H), 3.82 (s, 3H), 3.18 (q,
4H), 2.07 (p, 1H), 1.96 - 1.85 (m, 4H), 0.83 (d, 4H); LC-MS: m/z 323.12 (MH+).
Scheme 16, Cpd (Vl)*a Preparation: 5-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoic acid
Figure imgf000208_0002
To a solution of methyl 5-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate (175.0 mg, 0.540 mmol) in Methanol (6 mL) and THF (6 mL) a solution of lithium hydroxide hydrate (52.32 mg, 1.25 mmol) in 0.5 mL of water was added and the reaction mixture was stirred at 50 °C for 24 hours. After this time the reaction was cooled down, diluted with Water and washed with DCM 3 times. The aqueous phase was acidified with HCI 6 N until pH = 2 and then extracted three times with EtOAc. The organic portions were combined, washed with brine, dried over Na2SO4 and concentrated under high vacuum to afford the product of formula methyl 3-chloro-4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate, which was used in the next step without further purification.
Yield: 76 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.58 (s, 1H), 7.50 (s, 1H), 7.43 (s, 1H), 3.20 - 3.00 (m, 4H), 1.96
- 1.70 (m, 4H), 1.50 (tt, 1H), 0.95 - 0.43 (m, 4H); LC-MS: m/z 309.24 (MH+).
Scheme 16, Cpd (Vl)*b Preparation: 3-chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoic acid
Figure imgf000209_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 5- chloro-4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoic acid wherein methyl 3-chloro-4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate was used instead of methyl 5-chloro-4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate. The title compound was obtained in a yield of 80% (58 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 9.94 (s, 1H), 7.96 - 7.85 (m, 2H), 3.57 - 3.33 (m, 4H), 2.20 - 1.97
(m, 5H), 1.00 - 0.64 (m, 4H); LC-MS: m/z 309.24 (MH+).
Scheme 16, Cpd (Vll)*a Preparation: N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000209_0001
Example 95 To a solution of HATU (70.19 mg, 0.180 mmol), 5-chloro-4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoic acid (38.0 mg, 0.120 mmol) and N,N-Diisopropylethylamine (0.74 mL, 0.740 mmol) in DMF (1.5 mL), l-methyl-3-phenylpiperazine (26.03 mg, 0.150 mmol) was added. The reaction was stirred at RT for 30 hours. After this time a saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted three times with EtOAc. The organic portions were collected, washed with brine, residual water was removed by adding Na2SO4 and the solvent was evaporated under reduced pressure. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 35:65 of CH3CN/H2O +
0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted three times with AcOEt. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide as yellow powder.
Yield: 13 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.57 (d, 1H), 7.73 (d, 1H), 7.47 (d, 1H), 7.42 - 7.06 (m, 5H), 6.91
(s, 1H), 5.89 - 4.32 (m, 1H), 3.52 - 3.38 (m, 1H), 3.14 - 2.58 (m, 5H), 2.45 - 1.82 (m, 9H), 1.77 - 1.51
(m, 1H), 1.40 - 1.05 (m, 1H), 0.80 (d, 4H); LC-MS: m/z 467.48 (MH+).
Preparation: N-[2-chloro-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000210_0001
Example 96
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 95 N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 1,4-thiazinane 1,1-dioxide hydrochloride was used instead of l-methyl-3-phenylpiperazine. The title compound was obtained in a yield of 62% (32 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 9.54 (s, 1H), 7.38 (s, 1H), 7.24 (s, 1H), 4.46 (d, 1H), 3.80 (d,
1H), 3.64 - 3.51 (m, 2H), 3.39 (t, 1H), 3.17 (p, 3H), 3.05 (d, 3H), 2.70 (s, 1H), 2.10 - 1.96 (m, 1H), 1.87
(s, 4H), 0.80 (d, 4H); LC-MS: m/z 426.13 (MH+). Preparation: N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000211_0001
Example 97
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 95 N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-chloro-4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoic acid was used instead of 5-chloro-4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoic acid. The title compound was obtained in a yield of 68% (29 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 9.73 (d, 1H), 7.60 (dd, 2H), 7.54 - 7.10 (m, 5H), 7.02 - 5.75 (m,
1H), 5.75 - 4.65 (m, 1H), 3.07 - 2.95 (m, 3H), 2.82 (d, 1H), 2.63 (d, 1H), 2.36 - 2.10 (m, 4H), 2.05 - 1.74
(m, 8H), 0.82 (d, 4H); LC-MS: m/z 467.48 (MH+).
Preparation: N-[2-chloro-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000211_0002
Example 98
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 95 N-[2-chloro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)-5-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 3-chloro-4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoic acid was used instead of 5-chloro-4-(cyclopropanecarbonylamino)-2- pyrrolidin-l-ylbenzoic acid and 1,4-thiazinane 1,1-dioxide hydrochloride was used instead of 1- methyl-3-phenylpiperazine. The title compound was obtained in a yield of 31% (12 mg). 1H NMR (400 MHz, DMS0-d6) δ ppm 9.71 (s, 1H), 7.54 (d, 1H), 7.30 (d, 1H), 4.52 (d, 1H), 3.74 (d,
1H), 3.55 (t, 2H), 3.36 (t, 1H), 3.24 - 3.08 (m, 6H), 3.01 (t, 1H), 2.08 - 1.99 (m, 1H), 1.85 (s, 4H), 0.82
(d, 4H); LC-MS: m/z 426.13 (MH+).
Scheme 17
Figure imgf000212_0001
Step 1
Compound of formula II may be obtained from compound of formula I (synthesized according to the Scheme 1 7) by reaction with hydrazine hydrate in a suitable solvent such as EtOH, typically at
80 °C. The reaction takes about 3 days to complete.
Step 2
Compound of formula III may be obtained by cyclization of compound of formula II with triphosgene in presence of a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 1 hour to complete.
Step 3
Compound of formula IV may be obtained by activation of compound of formula III with a suitable phosphonium reagent, e.g. pyBOP, in presence of an organic base, e.g. DIPEA, followed by SnAr substitution, with a proper amine. The reaction is carried out in a suitable solvent, such as DMF, typically at room temperature. The reaction takes about 12 hours to complete.
Scheme 17, Cpd (ll) Preparation: N-[4-(hydrazinecarbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000212_0002
To a suspension of methyl 4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoate (2.0 g, 6.45 mmol) in Ethanol (10 mL), hydrazine hydrate (2.48 g, 32.25 mmol) was added and the reaction was stirred at 80 °C for 3 days. After this time, the reaction mixture was cooled down to RT and concentrated under vacuum. The residue was taken up with H2O and filtered. The solid was dried under vacuum affording the product of formula N-[4-(hydrazinecarbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide. The product was used in the next step without further purification.
Yield: 1.2 g
1H NMR (400 MHz, DMSO-d6) δ ppm 10.07 (s, 1H), 9.20 (s, 1H), 7.07 - 6.99 (m, 2H), 6.88 (dd, 1H),
4.32 (d, 2H), 3.19 - 3.09 (m, 4H), 1.88 - 1.82 (m, 4H), 1.78 - 1.69 (m, 1H), 0.77 (dq, 4H); LC-MS: m/z
289.25 (MH+).
Scheme 17, Cpd (III) Preparation: N-[4-(2-oxo-3H-l,3,4-oxadiazol-5-yl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000213_0001
To a suspension of N-[4-(hydrazinecarbonyl)-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
(0.5 g, 1.73 mmol) and N,N-Diisopropylethylamine (0.6 mL, 3.47 mmol) in DCM (35 mL), a solution of carbonic acid bis(trichloromethyl) ester (205.83 mg, 0.690 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at RT for 1 hour. After this time, MeOH was added and the mixture was concentrated under vacuum. The residue was taken up with DCM and filtered. The solid was washed with DCM and dried under vacuum affording the product of formula N-[4-(2-oxo-3H-l,3,4- oxadiazol-5-yl)-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide.
Yield: 300 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 12.33 (s, 1H), 10.25 (s, 1H), 7.36 - 7.12 (m, 2H), 7.01 (dd, 1H),
3.16 - 3.03 (m, 4H), 1.92 - 1.82 (m, 4H), 1.78 (dt, 1H), 0.87 - 0.71 (m, 4H); LC-MS: m/z 315.2 (MH+).
Scheme 17, Cpd (V)*a Preparation: N-[3-pyrrolidin-l-yl-4-(5-pyrrolidin-l-yl-l,3,4-oxadiazol-2- yl)phenyl]cyclopropanecarboxamide
Figure imgf000214_0001
Example 99
To a solution of N-[4-(2-oxo-3H-l,3,4-oxadiazol-5-yl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide (50.0 mg, 0.160 mmol) in DMF (1.5 mL), N,N-
Diisopropylethylamine (0.06 mL, 0.320 mmol) was added followed by l-benzotriazolyloxy-tris(l- pyrrolidinyl)phosphonium hexafluorophosphate (91.05 mg, 0.170 mmol) and pyrrolidine (0.03 mL,
0.320 mmol). The reaction mixture was stirred overnight at RT. The day after, the reaction mixture was directly purified by FC on reverse phase using basic conditions (from 5:95 of CH3CN/H2O + 10 mM ammonium bicarbonate aqueous solution adjusted to pH 10 with ammonia to 95: 5 of
CH3CN/H2O + 10 mM ammonium bicarbonate aqueous solution adjusted to pH 10 with ammonia).
Fractions containing the desired product were collected and concentrated under vacuum. The residue was again purified by FC on silica gel (eluting from DCM/EtOAc 8:2 to DCM/EtOAc 2:8) affording the product of formula N-[3-pyrrolidin-l-yl-4-(5-pyrrolidin-l-yl-l,3,4-oxadiazol-2- yl)phenyl]cyclopropanecarboxamide.
Yield: 8.1 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 10.22 (s, 1H), 7.34 - 7.17 (m, 2H), 7.00 (dd, 1H), 3.43 (d, 4H),
3.07 - 2.96 (m, 4H), 2.02 - 1.89 (m, 4H), 1.86 - 1.72 (m, 5H), 0.79 (dt, 4H); LC-MS: m/z 368.26 (MH+).
Preparation: N-[4-[5-(4,4-difluoropiperidin-l-yl)-l,3,4-oxadiazol-2-yl]-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide
Figure imgf000214_0002
Example 100 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 99 N-[3-pyrrolidin-l-yl-4-(5-pyrrolidin-l-yl-l,3,4-oxadiazol-2- yl)phenyl]cyclopropanecarboxamide wherein 4,4-difluoropiperidine was used instead of pyrrolidine. The title compound was obtained in a yield of 19% (13 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 10.24 (s, 1H), 7.32 - 7.23 (m, 2H), 7.01 (dd, 1H), 3.58 (t, 4H),
3.10 - 2.96 (m, 4H), 2.11 (dt, 4H), 1.81 (d, 5H), 0.80 (d, 4H); LC-MS: m/z 418.24 (MH+).
Scheme 18
Figure imgf000215_0001
Step 1
Compound of formula II may be obtained via Miyaura coupling between compound of formula I
(commercially available from Fluorochem) and Bis(pinacolato)diboron, in presence of a suitable catalyst, such as PdCl2(dppf)·CH2Cl2, a suitable base, e.g. AcOK, in a suitable solvent, e.g. 1,4 dioxane, typically at 100 °C. The reaction takes about 20 hours to complete.
Step 2
Compound of formula III may be obtained via Suzuki coupling between compound of formula II and the desired commercially available heteroaryl halide, in presence of a suitable palladium precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2- dimethoxyethane / water, typically at 95 °C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula IV may be obtained from compound of formula III by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes about 12 hours to complete. Step 4
Compound of formula V may be obtained by nitro reduction of compound of formula IV under catalytic hydrogenation conditions, using for example Pd/C in a suitable solvent such as MeOH, typically at room temperature. The reaction takes about 2 hours to complete.
Step 5
Compound of formula VI may be obtained by coupling reaction between compound of formula V and a proper commercially available acyl chloride, e.g. acetyl chloride, in presence of an organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 2 hours to complete.
Scheme 18. Cpd (II) Preparation: 2-(2-fluoro-4-nitrophenyl)-4,4,5,5-tetramethyl-l,3,2- dioxaborolane
Figure imgf000216_0001
4-bromo-3-fluoronitrobenzene (1.6 g, 7.27 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-l,3,2-dioxaborolane (1.94 g, 7.64 mmol), potassium acetate (2.16 g, 21.82 mmol) and [l,l'-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll), complex with dichloromethane (119.08 mg, 0.150 mmol) were suspended in 1,4-Dioxane (70 mL). The mixture was degassed through N2-vacuum cycles and then stirred under N2 atmosphere at 100 °C for 20h.
After this time, the mixture was filtered through a celite pad, washed with EtOAc and concentrated under vacuum affording the product of formula 2-(2-fluoro-4-nitrophenyl)-4,4,5,5-tetramethyl-
1,3,2-dioxaborolane, which was used in the next step without further purification.
Yield: 2.76 g
1H NMR (400 MHz, CDCI3) δ ppm 8.02 (dd, 1H), 7.97 - 7.84 (m, 2H), 1.40 (s, 12H).
Scheme 18, Cpd (III) Preparation: 2-(2-fluoro-4-nitrophenyl)-4-pyrrolidin-l-ylpyrimidine
Figure imgf000217_0001
In a proper vial 2-chloro-4-pyrrolidin-l-ylpyrimidine (250.0 mg, 1.36 mmol), 2-(2-fluoro-4- nitrophenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (0.62 g, 1.63 mmol), X-phos aminobiphenyl palladium chloride precatalyst (106.98 mg, 0.140 mmol) and tripotassium phosphate (879.26 mg,
4.08 mmol) were suspended in Water (2 mL), and 1,2-dimethoxyethane (15 mL). The vial was sealed and the resulting suspension was degassed under nitrogen flux for 5 min and then heated to
95 °C for 12h. After this time, the reaction mixture was cooled down to room temperature and diluted with water and extracted three times with EtOAc. The organic portions were combined, washed with brine, dried over Na2SO4, filtered and the solvent removed under vacuum affording the product of formula 2-(2-fluoro-4-nitrophenyl)-4-pyrrolidin-l-ylpyrimidine, which was used in the next step without further purification.
Yield: 570 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.32 (d, 1H), 8.27 - 8.05 (m, 3H), 6.54 (d, 1H), 3.71 - 3.38 (m,
4H), 1.99 (d, 4H); LC-MS: m/z 289.18 (MH+).
Scheme 18, Cpd (IV) Preparation: 2-(4-nitro-2-pyrrolidin-l-ylphenyl)-4-pyrrolidin-l-ylpyrimidine
Figure imgf000217_0002
To a stirred solution of 2-(2-fluoro-4-nitrophenyl)-4-pyrrolidin-l-ylpyrimidine (275.0 mg, 0.670 mmol) in DMSO (4 mL), pyrrolidine (0.17 mL, 2 mmol) was added and the mixture was stirred overnight at 120 °C. The day after, the reaction was cooled down to RT, diluted with water and the aqueous phase extracted three times with EtOAc. The combined organic portions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of cHex to AcOEt/cHex 70:30) affording the product of formula 2-
(4-nitro-2-pyrrolidin-l-ylphenyl)-4-pyrrolidin-l-yl pyrimidine.
Yield: 140 mg 1H NMR (400 MHz, DMS0-d6) δ ppm 8.22 (d, 1H), 7.47 (d, 3H), 6.42 (d, 1H), 3.31 (s, 4H), 3.03 (d,
4H), 1.94 (d, 4H), 1.78 (d, 4H); LC-MS: m/z 340.2 (MH+).
Scheme 18, Cpd (V) Preparation: 3-pyrrolidin-l-yl-4-(4-pyrrolidin-l-ylpyrimidin-2-yl)aniline
Figure imgf000218_0002
To a solution of 2-(4-nitro-2-pyrrolidin-l-ylphenyl)-4-pyrrolidin-l-ylpyrimidine (140.0 mg, 0.410 mmol) in MeOH, Pd/C 10% (43.9 mg, 0.040 mmol) was added. The reaction mixture was stirred for
2 hours under H2 atmosphere at room temperature. After this time, the mixture was filtered through a celite pad, washed with MeOH and concentrated under vacuum affording the product of formula 3-pyrrolidin-l-yl-4-(4-pyrrolidin-l-ylpyrimidin-2-yl)aniline, which was used in the next step without further purification.
Yield: 142 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 8.11 (d, 1H), 7.12 (d, 1H), 6.20 (d, 1H), 6.06 - 5.85 (m, 2H), 4.99
(s, 2H), 3.49 - 3.10 (m, 4H), 3.17 - 2.75 (m, 4H), 1.93 (t, 4H), 1.80 - 1.67 (m, 4H); LC-MS: m/z 310.26
(MH+).
Scheme 18. Cpd (VI) Preparation: N-[3-pyrrolidin-l-yl-4-(4-pyrrolidin-l-ylpyrimidin-2- yl)phenyl]acetamide
Figure imgf000218_0001
Example 101
To a mixture of N,N-Diisopropylethylamine (119.91 uL, 0.690 mmol) and 3-pyrrolidin-l-yl-4-(4- pyrrolidin-l-ylpyrimidin-2-yl)aniline (142.0 mg, 0.460 mmol) in DCM (4.494 mL) at 0 °C, acetyl chloride (30 uL, 0.410 mmol) was added dropwise and the mixture was stirred at RT for 2 hours.
After this time, MeOH was added and the reaction mixture was concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O
+ 0.1% of HCOOH to 40:60 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted three times with EtOAc. The combined organic portions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula N-[3-pyrrolidin-l-yl-4-(4- pyrrolidin-l-ylpyrimidin-2-yl)phenyl]acetamide.
Yield: 22 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 9.80 (s, 1H), 8.16 (d, 1H), 7.23 (d, 1H), 7.05 (d, 1H), 6.95 (dd,
1H), 6.30 (d, 1H), 3.42 (s, 4H), 3.00 - 2.89 (m, 4H), 2.04 (s, 3H), 1.94 (s, 4H), 1.80 - 1.68 (m, 4H); LC-
MS: m/z 352.46 (MH+).
Scheme 19
Figure imgf000219_0001
Step 1
Compound of formula II may be obtained by SNAr substitution between compound of formula I and benzyl mercaptan, in presence of a suitable base, e.g. K2CO3, in a suitable aprotic solvent, e.g. DMF, typically at RT. The reaction takes about 20 hours to complete.
Step 2
Compound of formula III may be obtained by oxidative chlorination of compound of formula II to the corresponding sulphonyl chloride in the presence of a suitable oxidizing agent, e.g. dichloro-5,5- dimethylimidazolidine-2, 4-dione, followed by treatment with a proper amine. The reaction is carried out in a suitable solvent, e.g. DCM, at a temperature between 0 °C and room temperature.
The reaction takes about 16 hours to complete Step 3
Compound of formula IV may be obtained from compound of formula III by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes about 12 hours to complete.
Step 4
Compound of formula V may be obtained by nitro reduction of compound of formula IV in the presence of Tin (III) chloride dehydrate in a suitable solvent, e.g. EtOH, typically at 80 °C. The reaction takes about 1 hour to complete.
Step 5
Compound of formula VI may be obtained by coupling reaction between compound of formula V and a proper commercially available acyl chloride, e.g. cyclopropanecarbonyl chloride, in presence of an organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 12 hours to complete.
Scheme 19, Cpd (II) Preparation: l-benzylsulfanyl-2-fluoro-4-nitrobenzene
Figure imgf000220_0001
To a solution of l,2-difluoro-4-nitrobenzene (2.0 g, 12.57 mmol) in DMF (13.5 mL), phenylmethanethiol (1.48 mL, 12.57 mmol) was added followed by potassium carbonate
(1.91 g, 13.83 mmol). The reaction was stirred at room temperature overnight. After this time the reaction was diluted with H2O and filtered. The solid was washed with H2O and dried under vacuum affording the product of formula l-benzylsulfanyl-2-fluoro-4-nitrobenzene (N0806-30-1: 3.3 g,
12.53 mmol, 99.7% yield) which was used in the next step without further purification.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.13 - 8.03 (m, 2H), 7.72 (t, 1H), 7.46 (d, 2H), 7.39 - 7.24 (m,
3H)
4.47 (s, 2H); LC-MS: m/z 264.01 (MH+).
Scheme 19, Cpd (lll)*a Preparation: l-(2-fluoro-4-nitrophenyl)sulfonyl-4-methylpiperazine
Figure imgf000221_0001
To a suspension of l-benzylsulfanyl-2-fluoro-4-nitrobenzene (100.0 mg, 0.380 mmol) in a mixture of MeCN (3.8 mL), Water (0.100 mL) and acetic acid (49.0 uL, 0.860 mmol) was added 1,3-dichloro-
5, 5-dimethylimidazolidine-2, 4-dione (149.66 mg, 0.760 mmol) at 0 °C. After stirring for 2h at 0 °C, the reaction mixture was concentrated under reduced pressure, the residue was dissolved in DCM
(1.5 mL) and again cooled to 0 °C. Then a saturated solution of NaHCO3 was added and the mixture was stirred for 15 minutes at 0 °C. Afterwards the mixture was extracted with DCM (X2) and the combined organic layers were washed with brine, dried over Na2SO4, filtered and evaporated under reduced pressure. The residue was dissolved in DCM (1.5 mL) and l-methyl-3-phenylpiperazine
(66.95 mg, 0.380 mmol) was added at 0 °C followed by l,4-diazabicyclo[2.2.2]octane (51.12 mg,
0.460 mmol) and the mixture was allowed to warm to room temperature. After stirring 16 h, the reaction mixture was quenched with water and extracted with DCM (x3). The organic layers were washed with brine, dried using a phase separator and concentrated to afford a crude that was purified by FC on silica gel (eluting from 100% of cHex to cHex/EtOAc 80:20) affording the product of formula l-(2-fluoro-4-nitrophenyl)sulfonyl-4-methyl-2-phenylpiperazine.
Yield: 130 mg
1H NMR (400 MHz, DMSO-d6) δ 8.37 (dd, 1H), 8.24 - 8.17 (m, 1H), 8.15 - 8.05 (m, 1H), 7.47 - 7.43 (m,
2H), 7.36 - 7.18 (m, 3H), 5.09 - 5.02 (m, 1H), 3.75 - 3.66 (m, 1H), 3.31 - 3.18 (m, 2H), 2.77 - 2.62 (m,
1H), 2.20 - 2.07 (m, 4H), 1.79 (td, 1H); LC-MS: m/z 380.12 (MH+).
Scheme 19, Cpd (lll)*b Preparation: l-(2-fluoro-4-nitrophenyl)sulfonyl-4-methylpiperazine
Figure imgf000221_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 1-
(2-fluoro-4-nitrophenyl)sulfonyl-4-methylpiperazine wherein 1 -methylpiperazine was used instead of l-methyl-3-phenylpiperazine. The title compound was obtained in a quantitative yield (700 mg). 1H NMR (400 MHz, DMSO-d6) δ 8.41 (dd, 1H), 8.28 - 8.22 (m, 1H), 8.08 - 8.02 (m, 1H), 3.15 (t, 4H),
2.37 (t, 4H), 2.17 (s, 3H); LC-MS: m/z 304.05 (MH+).
Scheme 19, Cpd (IV)*a Preparation: 4-methyl-l-(4-nitro-2-pyrrolidin-l-ylphenyl)sulfonyl-2- phenylpiperazine
Figure imgf000222_0001
A mixture of pyrrolidine (0.56 g, 7.91 mmol) and l-(2-fluoro-4-nitrophenyl)sulfonyl-4-methyl-2- phenylpiperazine (1.0 g, 2.64 mmol) in DMSO (15 mL) was heated to 120 °C for 12 h. After this time the reaction was diluted with water and then extracted with AcOEt x3. Organic phase was dried over Na2SO4, filtered and concentrated under vacuum. The crude was purified by FC on silica gel
(eluting from 100% of cHex 100% to cHex/EtOAc 80:20) to afford the product of formula 4-methyl- l-(4-nitro-2-pyrrolidin-l-ylphenyl)sulfonyl-2-phenylpiperazine.
Yield: 397 mg
LC-MS: m/z 431.19 (MH+).
Scheme 19, Cpd (IV)*b Preparation: l-methyl-4-(4-nitro-2-pyrrolidin-l-ylphenyl)sulfonylpiperazine
Figure imgf000222_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4- methyl-l-(4-nitro-2-pyrrolidin-l-ylphenyl)sulfonyl-2-phenylpiperazine wherein l-(2-fluoro-4- nitrophenyl)sulfonyl-4-methylpiperazine was used instead of l-(2-fluoro-4-nitrophenyl)sulfonyl-4- methyl-2-phenylpiperazine. The title compound was obtained in yield of 70% (490 mg).
LC-MS: m/z 355.15 (MH+).
Scheme 19, Cpd (V)*a Preparation: 4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l- ylaniline
Figure imgf000223_0001
To a solution of 4-methyl-l-(4-nitro-2-pyrrolidin-l-ylphenyl)sulfonyl-2-phenylpiperazine (397.0 mg,
0.920 mmol) in Ethanol (10 mL), tin(lll) chloride dihydrate (1.3 g, 5.53 mmol) was added and the reaction was stirred at 80 °C for lh. After this time EtOH was evaporated under reduced pressure and the residue dissolved in EtOAc. A precipitate was formed and it was filtered on Hirsch funnel under reduced pressure. The filtrate was evaporated under reduced pressure and then purified by
SCX first washing with MeOH and then eluting with NH3 IN in MeOH. Basic fractions were collected and concentrated under vacuum affording the product of formula 4-(4-methyl-2-phenylpiperazin- l-yl)sulfonyl-3-pyrrolidin-l-ylaniline. The product was used in the next step without further purification.
Yield: 200 mg
LC-MS: m/z 401.27 (MH+).
Scheme 19, Cpd (V)*b Preparation: 4-(4-methylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l-ylaniline
Figure imgf000223_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound 4-
(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l-ylaniline wherein l-methyl-4-(4-nitro-2- pyrrolidin-l-ylphenyl)sulfonylpiperazine was used instead of 4-methyl-l-(4-nitro-2-pyrrolidin-l- ylphenyl)sulfonyl-2-phenylpiperazine. The title compound was obtained in yield of 9% (40 mg).
LC-MS: m/z 323.18 (MH+).
Scheme 19, Cpd (Vl)*a Preparation: N-[4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000224_0001
Example 102
To a solution of 4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l-ylaniline (199.0 mg,
0.500 mmol) in DCM (6.387 mL), N,N-Diisopropylethylamine (0.13 mL, 0.750 mmol) was added followed by cyclopropanecarbonyl chloride (0.06 mL, 0.650 mmol) . The reaction was stirred at
RT/ON. The day after the reaction was diluted with DCM, washed with a s.s. of NaHCO3, brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on RP using basic conditions (eluting from 100% of water + 0.1% of NH4OH to 20:80 of CH3CN/water + 0.1% of
NH4OH) to afford the product of formula N-[4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide.
Yield: 72 mg
1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 7.75 (d, 1H), 7.66 (d, 1H), 7.43 - 7.35 (m, 2H), 7.32 -
7.12 (m, 4H), 5.01 - 4.93 (m, 1H), 3.57 - 3.47 (m, 1H), 3.26 - 2.91 (m, 8H), 2.63 - 2.57 (m, 1H), 2.19
(dd, 1H), 1.90 - 1.65 (m, 6H), 0.91 - 0.67 (m, 5H); LC-MS: m/z 467.21 (MH+).
Preparation: N-[4-(4-methylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide
Figure imgf000224_0002
Example 103
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 102 N-[4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide wherein 4-(4-methylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l- ylaniline was used instead of 4-(4-methyl-2-phenylpiperazin-l-yl)sulfonyl-3-pyrrolidin-l-ylaniline.
The title compound was obtained in yield of 73% (33 mg). 1H NMR (400 MHz, DMS0-d6) δ ppm 10.52 - 10.43 (m, 1H), 7.72 - 7.61 (m, 2 H), 7.26 (d, 1H), 3.24 -
3.13 (m, 4H), 3.06 - 2.95 (m, 3H), 2.37 - 2.21 (m, 5H), 2.18 - 2.10 (m, 3H), 1.93 - 1.73 (m, 5H), 0.83
(d, 4H); LC-MS: m/z 393.18 (MH+).
Scheme 20
Figure imgf000225_0002
Step 1
Compound of formula II may be obtained from compound of formula I (synthesized according to the Scheme 1) by removing the Boc protecting group under acidic conditions, e.g. TFA, typically at
RT. The reaction takes from about 3 hours to 12 hours to complete.
Step 2
Compound of formula III may be obtained via sulphonamide formation between compound of formula II and methanesulfonyl chloride in presence of a suitable organic base, e.g. TEA, in a suitable solvent, e.g. DCM, typically at RT. The reaction takes about 5 minutes to complete
Scheme 20, Cpd (ll)*a Preparation: N-[4-[4-(methylamino)piperidine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000225_0001
To a solution of tert-butyl N-[l-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperidin-
4-yl]-N-methylcarbamate (160.0 mg, 0.340 mmol) in DCM (5.128 mL), at RT, Trifluoroacetic acid
(0.26 mL, 3.4 mmol) was added and the resulting reaction mixture was stirred at RT overnight. The mixture was concentrated under vacuum, dissolved in ImL of MeOH and purified by SCX first washing with MeOH and then eluting with NH3 IN in MeOH. Basic fractions were collected and concentrated under vacuum affording the product of formula N-[4-[4-(methylamino)piperidine-l- carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide, which was used in the next step without further purification.
Yield: 86 mg
LC-MS: m/z 371.28 (MH+).
Scheme 20, Cpd (Il)*b Preparation: N-[4-(4-aminopiperidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000226_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[4-[4-(methylamino)piperidine-l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide wherein tert-butyl N-[l-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperidin-4- yl)carbamate was used instead of tert-butyl N-[l-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l- ylbenzoyl]piperidin-4-yl]-N-methylcarbamate. The title compound was obtained in a yield 70% (86 mg).
LC-MS: m/z 457.48 (MH+)
Scheme 20, Cpd (lll)*a Preparation: N-[4-[4-[methyl(methylsulfonyl)amino]piperidine-l-carbonyl]-
3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000226_0002
Example 104
To a 0 °C cooled solution of N-[4-[4-(methylamino)piperidine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide (86.0 mg, 0.230 mmol) and triethylamine (0.05 mL, 0.350 mmol) in DCM (3.479 mL), methanesulfonyl chloride (0.02 mL, 0.280 mmol) was added. The mixture was warmed at room temperature and stirred for 5 minutes. The mixture was diluted with DCM and washed with brine. The organic layer was filtered through a phase separator and concentrated under reduced pressure. The residue was purified by FC on RP using acid conditions (eluting from
5:95 of CH3CN/H2O + 0.1% of HCOOH to 35:65 of CH3CN/H2O + 0.1% of HCOOH) to give the product of formula N-[4-[4-[methyl(methylsulfonyl)amino]piperidine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide.
Yield: 33 mg
1H NMR (400 MHz, DMSO-d6) δ 10.09 (d, 1H), 7.17 - 6.79 (m, 3H), 4.70 - 4.50 (m, 1H), 3.85 - 3.77
(m, 1H), 3.62 - 3.49 (m, 1H), 3.23 - 2.82 (m, 8H), 2.68 (d, 4H), 1.94 - 1.48 (m, 9H),0.78 (dd, 4H); LC-
MS: m/z 449.26 (MH+).
Preparation: N-[4-[4-(methanesulfonamido)piperidine-l-carbonyl]-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide
Figure imgf000227_0001
Example 105
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[4-[4-(methylamino)piperidine-l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide wherein N-[4-(4-aminopiperidine-l-carbonyl)-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-[4-[4-(methylamino)piperidine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide. The title compound was obtained in a yield 33% (35 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.09 (s, 1H), 7.16 (dd, 1H), 7.09 -7.02 (m, 1H), 6.96 -6.83 (m, 2H),
4.31 (dd, 1H), 3.53 -3.35 (m, 2H), 3.21 - 2.78 (m, 9H), 1.87 (dd, 5H), 1.76 (h, 2H), 1.33 (dd, 2H), 0.77
(dt, 4H); LC-MS: m/z 435.23 (MH+). Scheme 21
Figure imgf000228_0001
Step 1
Compound of formula II may be obtained from compound of formula I (synthesized according to the Scheme 1) by removing the Boc protecting group under acidic conditions, e.g. TFA, typically at
RT. The reaction takes about 2 hours to complete.
Step 2
Compound of formula III may be obtained via reductive amination between compound of formula
II and a proper aldehyde, e.g. formaldehyde, in the presence of a suitable reducing agent, e.g. sodium triacetoxyborohydride. The reaction is carried out in a suitable solvent, e.g. MeOH, typically at RT. The reaction takes about 12 hours to complete.
Scheme 21, Cpd (ll)*a Preparation: N-[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine- l-carbonyl]phenyl]cyclopropanecarboxamide
Figure imgf000228_0002
To a solution of tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate (24.3 mg, 0.041 mmol) in DCM (2 mL), trifluoroacetic acid (0.095 mL, 1.243 mmol) was added. The reaction mixture was stirred at RT for 2 hour. After this time the mixture was concentrated to dryness under vacuum. The residue was purified by SCX first washing with MeOH and then eluting with NH3 1N in MeOH. Basic fractions were collected and concentrated under vacuum affording the product of formula N-[3-pyrrolidin-l- yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]phenyl]cyclopropanecarboxamide as a racemic mixture. The product was used in the next step without further purification.
Yield: 19 mg
LC-MS: m/z 487.18 (MH+). Scheme 21, Cpd (ll)*b Preparation: N-[4-[2-(3-cyanophenyl)piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000229_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl)phenyl)cyclopropanecarboxamide wherein tert-butyl 3-(3-cyanophenyl)-4-[4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperazine-l-carboxylate was used instead of tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate. The title compound was obtained as a racemic mixture in a yield 97% (44 mg).
Scheme 21, Cpd (ll)*c Preparation: N-[4-[2-(4,5-dimethyl-l,3-thiazol-2-yl)piperazine-l-carbonyl]-
3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000229_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein tert-butyl 4-[4-(cyclopropanecarbonylamino)-
2-pyrrolidin-l-ylbenzoyl]-3-(4,5-dimethyl-l,3-thiazol-2-yl)piperazine-l-carboxylate was used instead of tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate. The title compound was obtained as a racemic mixture in a quantitative yield (59 mg).
LC-MS: m/z 454.24 (MH+). Scheme 21, Cpd (ll)*d Preparation: N-[4-[2-(4-methylphenyl)piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000230_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein tert-butyl 4-[4-(cyclopropanecarbonylamino)-
2-pyrrolidin-l-ylbenzoyl]-3-(4-methylphenyl)piperazine-l-carboxylate was used instead of tertbutyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate. The title compound was obtained as a racemic mixture in a yield of 46% (36 mg).
LC-MS: m/z 433.38 (MH+).
Scheme 21, Cpd (ll)*e Preparation: N-[4-[2-(3-chloro-5-methylphenyl)piperazine-l-carbonyl]-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000230_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein tert-butyl 3-(3-chloro-5-methylphenyl)-4-[4-
(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]piperazine-l-carboxylate was used instead of tert-butyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate. The title compound was obtained as a racemic mixture in a yield of 63% (68 mg).
LC-MS: m/z 467.25 (MH+). Scheme 21, Cpd (ll)*f Preparation: N-[4-[2-(3-methylphenyl)piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000231_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein tert-butyl 4-[4-(cyclopropanecarbonylamino)-
2-pyrrolidin-l-ylbenzoyl]-3-(3-methylphenyl)piperazine-l-carboxylate was used instead of tertbutyl 4-[4-(cyclopropanecarbonylamino)-2-pyrrolidin-l-ylbenzoyl]-3-[2-
(trifluoromethyl)phenyl]piperazine-l-carboxylate. The title compound was obtained as a racemic mixture in a yield of 90% (118 mg).
LC-MS: m/z 433.31 (MH+).
Scheme 21, Cpd (lll)*a Preparation: N-[4-[4-methyl-2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide
Figure imgf000231_0002
Example 106
To a solution of N-[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide (19.0 mg, 0.040 mmol) in Methanol (1.000 mL) formaldehyde (0.03 mL, 0.390 mmol) was added followed by sodium triacetoxyborohydride(99.66 mg, 0.234 mmol). The reaction was stirred at room temperature ON. The day after the reaction was diluted with EtOAc and washed with a saturated solution of NaHCO3. The organic phase was dried using a phase separator and concentrated. The crude was purified by FC on silica gel (eluting from cHex/EtOAc 50:50 to cHex/EtOAc 20:80 ), obtaining the product of formula N-[4-[4-methyl-2- [2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide as a racemic mixture.
Yield: 6.5 mg
NMR (400 MHz, DMSO-d6) δ 10.15 -10.02 (m, 1H), 7.99 -7.77 (m, 1H), 7.77 -7.58 (m, 2H), 7.56 -
7.44 (m, 1H), 7.13 -7.06 (m, 1H), 7.00 -6.97 (m, 1H), 6.91 -6.61 (m, 1H), 5.84 -5.32 (m, 1H), 3.88 -
3.60 (m, 2H), 3.26 -3.05 (m, 3H), 2.86 -2.72 (m, 3H), 2.47 -2.38 (m, 1H), 2.24 -2.05 (m, 4H), 2.05 -
1.84 (m, 3H), 1.82 -1.57 (m, 2H), 0.81 -0.67 (m, 4H); LC-MS: m/z 501.31 (MH+).
Preparation: N-[4-[2-(3-cyanophenyl)-4-methylpiperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000232_0001
Example 107
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein N-[4-[2-(3-cyanophenyl)piperazine-l- carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-[3-pyrrolidin-l- yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]phenyl]cyclopropanecarboxamide. The title compound was obtained as a racemic mixture in a yield 35% (16 mg).
1H NMR (400 MHz, DMSO-d6) δ 10.11 (s, 1H), 8.16 -7.86 (m, 1H), 7.82 -7.37 (m, 3H), 7.21 -6.67 (m,
3H), 5.92 -5.54 (m, 1H), 3.44 (dd, 2H), 3.24 -2.83 (m, 4H), 2.78 -2.59 (m, 2H), 2.40 -2.10 (m, 4H),
2.09 -1.86 (m, 3H), 1.83 -1.57 (m, 3H), 0.82 -0.62 (m, 4H); LC-MS: m/z 458.29 (MH+).
Preparation: N-[4-[2-(4,5-dimethyl-l,3-thiazol-2-yl)-4-methylpiperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000233_0001
Example 108
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein N-[4-[2-(4,5-dimethyl-l,3-thiazol-2- yl)piperazine-l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide. The title compound was obtained as a racemic mixture in a yield 89% (54 mg).
NMR (400 MHz, DMSO-d6) δ 10.15 -10.06 (m, 1H), 7.21 -6.77 (m, 3H), 5.90 -4.26 (m, 1H), 3.61 -
3.35 (m, 2H), 3.30 -2.56 (m, 6H), 2.39 -2.12 (m, 10H), 2.08 -1.61 (m, 6H), 0.89 - 0.70 (d, 4H); LC-MS m/z 468.49 (MH+).
Preparation: N-[4-[4-methyl-2-(4-methylphenyl)piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000233_0002
Example 109
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein N-[4-[2-(4-methylphenyl)piperazine-l- carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-[3-pyrrolidin-l- yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]phenyl]cyclopropanecarboxamide. The title compound was obtained as a racemic mixture in a yield 32% (11 mg). 1H NMR (400 MHz, DMSO-d6) δ 10.21 -9.95 (m, 1H), 7.66 -7.23 (m, 2H), 7.22 -6.69 (m, 5H), 5.84 -
4.25 (m, 1H), 3.46 -3.38 (m, 1H), 3.27 -2.55 (m, 6H), 2.46 -2.08 (m, 7H), 2.06 -1.85 (m, 4H), 1.81 -
1.53 (m, 3H), 0.93 -0.65 (m, 4H); LC-MS: m/z 447.30 (MH+).
Preparation: N-[4-[2-(3-chloro-5-methylphenyl)-4-methylpiperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000234_0001
Example 110
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein N-[4-[2-(3-chloro-5-methylphenyl)piperazine- l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-[3-pyrrolidin- l-y|-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]phenyl]cyclopropanecarboxamide. The title compound was obtained as a racemic mixture in a yield 60% (41 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 10.10 (s, 1H), 7.59 - 6.76 (m, 6H), 5.79 - 5.64 (m, 1 H), 4.72 -
4.35 (m, 1 H), 3.48 - 2.60 (m, 7 H), 2.40 - 2.13 (m, 7 H), 2.08 - 1.63 (m, 6 H), 0.85 - 0.69 (m, 4 H); LC-
MS: m/z 481.28 (MH+)
The racemic mixture (Example 110) was then separated into single enantiomers by chiral semi- preparative HPLC
Column Chiralpak IC (25 x 3.0 cm), 5 μ
Mobile phase n-Hexane/(Ethanol + 0.1% I PA)
Preparative chiral 80/20 % v/v chromatography Flow rate (ml/min) 37 ml/min protocol: DAD detection 220 nm
Loop 1000 μL
Total amount 39 mg
Injection 11.1 mg (each injection)
Enantiomer 1 Yield: Rt 10.84 min 100% ee 16
LC-MS: m/z 481.28 (MH+)
N-[4-[2S or 2R-(3-chloro-5-methylphenyl)-4- methyl piperazine-l-carbonyl]-3-pyrrolidin-l-
Figure imgf000235_0001
ylphenyl)cyclopropanecarboxamide
2R or 2S enantiomer
Example 110a
Enantiomer 2 Yield: Rt 14.49 min 100% ee
LC-MS: m/z 481.28 (MH+)
N-[4-[2S or 2R-(3-chloro-5-methylphenyl)-4- methylpiperazine-l-carbonyl]-3-pyrrolidin-l-
Figure imgf000235_0002
ylphenyl)cyclopropanecarboxamide
2R or 2S enantiomer Example 110b
Preparation of N-[4-[2S or 2R-(3-chloro-5-methylphenyl)-4-methylpiperazine-l-carbonyl]-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide hydrochloride
Figure imgf000236_0001
Example 110a
N-[4-[2S or 2R -(3-chloro-5-methylphenyl)-4-methylpiperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide (Enantiomer 1) (15.7 mg, 0.033 mmol) was dissolved in Methanol dry (1 mL) and treated with 1 eq of HCI in dioxane (0.033 mL, 0.033 mmol) to afford, after evaporation, the product of formula N-[4-[2R or 2S-(3-chloro-5-methylphenyl)-4- methylpiperazine-l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide hydrochloride.
Yield: 16.1 mg
LC-MS: m/z 481.25 (MH+).
Preparation of N-[4-[2S or 2R-(3-chloro-5-methylphenyl)-4-methylpiperazine-l-carbonyl]-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide hydrochloride
Figure imgf000236_0002
Example 110b
N-[4-[2S or 2R -(3-chloro-5-methylphenyl)-4-methylpiperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide (Enantiomer 2) (14.9 mg, 0.031 mmol) was dissolved in Methanol dry (1 mL) and treated with 1 eq of HCI in dioxane (0.031 mL, 0.031 mmol) to afford, after evaporation, the product of formula N-[4-[2R or 2S-(3-chloro-5-methylphenyl)-4- methylpiperazine-l-carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide hydrochloride.
Yield: 13.7
LC-MS: m/z 481.25 (MH+).
Preparation: N-[4-[4-methyl-2-(3-methylphenyl)piperazine-l-carbonyl]-3-pyrrolidin-l- ylphenyl]cyclopropanecarboxamide
Figure imgf000237_0001
Example 111
The synthesis of the title compound was effected analogously to the synthesis of the compound N-
[3-pyrrolidin-l-yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l- carbonyl]phenyl]cyclopropanecarboxamide wherein N-[4-[2-(3-methylphenyl)piperazine-l- carbonyl]-3-pyrrolidin-l-ylphenyl]cyclopropanecarboxamide was used instead of N-[3-pyrrolidin-l- yl-4-[2-[2-(trifluoromethyl)phenyl]piperazine-l-carbonyl]phenyl]cyclopropanecarboxamide. The title compound was obtained as a racemic mixture in a yield 65% (79 mg).
NMR (400 MHz, DMSO-d6) δ 10.17 -10.03 (m, 1H), 7.54 -7.42 (m, 1H), 7.30 -7.12 (m, 2H), 7.11 -
6.92 (m, 3H), 6.87 -6.81 (m, 1H), 5.84 -4.30 (m, 1H), 3.50 -3.34 (m, 1H), 3.26 -2.59 (m, 6H), 2.40 -
2.11 (m, 7H), 2.05 -1.84 (m, 4H), 1.80 -1.58 (m, 3H), 0.95 -0.66 (m, 4H); LC-MS: m/z 447.29 (MH+)
Scheme 22
Figure imgf000237_0002
Step 1
Compound of formula II may be obtained from compound of formula I (commercially available from
Enamine) by coupling with 4-(trifluoromethyl)piperidine hydrochloride in presence of a suitable coupling agent, e.g. HATU, and a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes about 5 hours to complete. Step 2
Compound of formula III may be obtained from compound of formula II by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. CH3CN, typically at 80° C. The reaction takes about 12 hours to complete.
Step 3
Compound of formula IV may be obtained by nitrile hydrolysis of compound of formula III under standard literature conditions such as by reaction with NaOH in the presence of H2O2. The reaction is carried out in a suitable solvent, e.g. MeOH, typically at RT. The reaction takes about 12 hours to complete
Step 4
Compound of formula V may be obtained from compound of formula IV via N-formyl imide formation, by reaction with a suitable formilating agent such as 1,1-dimethoxy-N, N- dimethylmethanamine, followed by cyclization with NH2NH2. The reaction is carried out in a suitable solvents, e.g. DMF, typically at a temperature between RT and 120 °C.
Scheme 22, Cpd (II) Preparation: 3-fluoro-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile
Figure imgf000238_0001
To a solution of HATU (3454.12 mg, 9.08 mmol), 4-cyano-2-fluorobenzoic acid (1.5 g, 9.08 mmol) and 4-(trifluoromethyl)piperidine hydrochloride (1722.47 mg, 9.08 mmol) in DMF (30 mL) DIPEA
(4.85 mL, 27.25 mmol) was added. The reaction was stirred at RT for 5 h. After this time the reaction was diluted with H2O and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of cHex to AcOEt/cHex 1:1) affording the product of formula 3-fluoro-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile.
Yield: 2.5 g 1H NMR (400 MHz, DMSO-d6) δ 8.00 (dd, 1H), 7.81 (dd, 1H), 7.75 -7.62 (m, 1H), 4.60 (d, 1H), 3.41 (d,
1H), 3.14 (t, 1H), 2.85 (td, 1H), 2.75 -2.60 (m, 2H), 2.00 -1.90 (m, 1H), 1.85 -1.76 (m, 1H), 1.53 -1.28
(m, 1H); LC-MS: m/z 301.19 (MH+).
Scheme 22, Cpd (lll) Preparation: 3-pyrrolidin-l-yl-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzonitrile
Figure imgf000239_0001
To a solution of 3-fluoro-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile (1.5 g, 5 mmol) in MeCN (20 mL) pyrrolidine (1.67 mL, 19.98 mmol) was added and the reaction was stirred at 80
°C ON. After this time the reaction was cooled down to RT and H2O was added. The mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4 filtered and concentrated under vacuum. The residue was purified by FC on silica gel
(eluting from 100% of cHex to cHex/ AcOEt 3:7) affording the product of formula 3-pyrrolidin-l-yl-
4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile.
Yield: 1.6 g
1H NMR (400 MHz, DMSO-d6) δ 7.19 (dd, 1H), 7.10 -7.00 (m, 2H), 4.60 (t, 1H), 3.51 (dd, 1H), 3.21 -
2.56 (m, 7H), 1.97 -1.68 (m, 6H), 1.55 -1.24 (m, 2H); LC-MS: m/z 352.20 (MH+).
Scheme 22. Cpd (IV) Preparation: 3-pyrrolidin-l-yl-4-[4-(trifluoromethyl)piperidine-l- carbonyl)benzamide
Figure imgf000239_0002
To a solution of 3-pyrrolidin-l-yl-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile (285.37 mg, 0.810 mmol) in Methanol (3 mL) sodium hydroxide (0.27 mL, 0.810 mmol) was added and the reaction was stirred ON at RT. The day after hydrogen peroxide (0.2 mL, 1.97 mmol) was added followed by further sodium hydroxide (0.27 mL, 0.810 mmol) and the reaction was stirred at RT for 4h. After this time the reaction was diluted with H2O and extracted with AcOEt 3 times. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula 3-pyrrolidin-l-yl-4-[4-(trifluoromethyl)piperidine-l- carbonyl]benzamide. The product was used in the next step without further purification.
Yield: 120 mg
1H NMR (400 MHz, DMSO-d6) δ 7.24 -6.97 (m, 3H), 4.62 (t, 1H), 3.53 (t, 1H), 3.26 -2.52 (m, 7H),
1.96 -1.67 (m, 6H), 1.49 -1.27 (m, 2H); LC-MS: m/z 370.18 (MH+).
Scheme 22, Cpd (V) Preparation: [2-pyrrolidin-l-yl-4-(4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000240_0001
Example 112
To a solution of 3-pyrrolidin-l-yl-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzamide (120.0 mg,
0.320 mmol) in DMF (1.793 mL), l,l-dimethoxy-N,N-dimethylmethanamine (0.09 mL, 0.650 mmol) was added and the reaction was stirred for 1.5h at 120 °C. After this time the reaction was cooled down to RT and H2O was added. The reaction was with EtOAc (x3), washed with brine, dried on Na2SO4 and filtered and concentrated under vacuum. The residue was dissolved with Ethanol (5 mL) and hydrazine (0.5 mL, 5.52 mmol) was added. The reaction was stirred at RT for 2 h. After this time the reaction was diluted with H2O and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of DCM to DCM/MeOH 9:1) to give the product of formula [2- pyrrolidin-l-yl-4-(4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone.
Yield: 40 mg
1H NMR (400 MHz, DMSO-d6) δ 14.17 (s, 1H), 8.36 (s, 1H), 7.46 -7.29 (m, 2H), 7.12 (dd, 1H), 4.64 (t,
1H), 3.61 (t, 1H), 3.28 (s, 2H), 3.17 -2.94 (m, 2H), 2.90 -2.59 (m, 3H), 2.05 -1.72 (m, 6H), 1.53 -1.27
(m, 2H); LC-MS: m/z 394.22 (MH+). Scheme 23
Figure imgf000241_0001
Step 1
Compound of formula II may be obtained via acyl chloride formation of compound of formula I
(commercially available from Enamine) by treatment with a suitable chlorinating agent, such as
SOCl2, typically at 90 °C, followed by coupling with l-Methyl-3-phenylpiperazine (commercially available from Enamine) in the presence of a suitable organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 16 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by SNAr substitution with pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120 °C. The reaction takes about 3 hours to complete.
Step 3
Compound of formula IV may be obtained by nitro reduction of compound of formula III using Zn dust in a suitable solvent, e.g. AcOH, typically at room temperature. The reaction takes about 2 hours to complete.
Step 4
Compound of formula V may be obtained by cyclization of compound of formula IV with a proper aldehyde, e.g. cyclopropanecarboxaldehyde, in the presence of a suitable oxidating agent, e.g. oxone. The reaction is carried out in a suitable mixture of solvents, e.g. DMF/H2O typically at RT.
The reaction takes about 4 hours to complete.
Scheme 23, Cpd (II) Preparation: (4-amino-2-fluoro-5-nitrophenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000242_0002
A mixture of 4-amino-2-fluoro-5-nitrobenzoic acid (200.0 mg, 1 mmol) and thionyl dichloride (1.46 mL, 19.99 mmol) was heated to reflux for 4 hours. After this time the reaction was cooled down to
RT and then concentrated under vacuum. The mixture was stripped three times with cyclohexane in order to remove thionyl dichloride in excess. The residue was suspended in DCM (4 mL) and 1- methyl-3-phenylpiperazine (176.15 mg, 1 mmol) was added followed by TEA (0.42 mL, 3 mmol) and the reaction was stirred overnight at RT. After this time the mixture was diluted with DCM and washed with a s.s. of NaHCO3 3 times. The aqueous phases were collected together and counterextracted with DCM. The combined organic phases were dried over a phase separator and concentrated under vacuum, affording the product of formula (4-amino-2-fluoro-5-nitrophenyl)-(4- methyl-2-phenylpiperazin-l-yl)methanone. The product was used in the next step without further purification.
Yield: 380 mg
LC-MS: m/z 359.36 (MH+).
Scheme 23, Cpd (Ill) Preparation: (4-amino-5-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000242_0001
To a solution of (4-amino-2-fluoro-5-nitrophenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone
(386.0 mg, 1.08 mmol) in DMSO (8 mL) pyrrolidine (0.27 mL, 3.23 mmol) was added. The mixture was stirred at 120°C for 2 h. After this time the mixture was cooled down to RT and diluted with H2O. The mixture was extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from cHex/AcOEt 8:2 to cHex/AcOEt 3:7) affording the product of formula
(4-amino-5-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone.
Yield: 230 mg 1H NMR (400 MHz, DMSO-d6) δ 7.77 -7.70 (m, 1H), 7.59 -7.44 (m, 1H), 7.44 -7.15 (m, 5H), 6.11 -
4.31 (m, 1H), 3.63 -3.31 (m, 3H), 3.20 -2.98 (m, 2H), 2.88 -2.76 (m, 1H), 2.74 -2.66 (m, 1H), 2.45 -
2.11 (m, 5H), 2.10 -1.85 (m, 3H), 1.82 -1.59 (m, 2H); LC-MS: m/z 410.22 (MH+).
Scheme 23, Cpd (IV) Preparation: (4,5-diamino-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone
Figure imgf000243_0001
To a solution of (4-amino-5-nitro-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l- yl)methanone (232.3 mg, 0.570 mmol) in AcOH (8 mL) zinc (370.9 mg, 5.67 mmol) was added and the reaction was stirred at RT for 2 hours. After this time the mixture was filtered and concentrated to residue by rotavapor. The crude was then purified by SCX first washing with MeOH and then eluting with NH3 IN in MeOH. Basic fractions were collected and concentrated under vacuum affording the product of formula (4,5-diamino-2-pyrrolidin-l-ylphenyl)-(4-methyl-2- phenylpiperazin-l-yl)methanone. The product was used in the next step without further purification.
Yield: 190 mg
1H NMR (400 MHz, DMSO-d6) δ 7.63 (d, 1H), 7.53 -7.10 (m, 5H), 6.49 -5.58 (m, 2H), 4.92 -3.97 (m,
2H), 3.23 -2.60 (m, 7H), 2.29 -2.13 (m, 5H), 1.88 (d, 5H), 1.62 (d, 2H); LC-MS: m/z m/z 381.06 (M+2H). Scheme 23, Cpd (V)*a Preparation: (2-cyclopropyl-6-pyrrolidin-l-yl-3H-benzimidazol-5-yl)-(4- methyl-2-phenylpiperazin-l-yl)methanone
Figure imgf000244_0001
Example 113
To a mixture of (4,5-diamino-2-pyrrolidin-l-ylphenyl)-(4-methyl-2-phenylpiperazin-l-yl)methanone
(90.0 mg, 0.240 mmol) and cyclopropanecarboxaldehyde (24.93 mg, 0.360 mmol) in DMF (1.5 mL)/Water (0.250 mL) Oxone (87.48 mg, 0.140 mmol) was added. The reaction was stirred at RT for
4 h. After this time a s.s. of NaHCO3 was added and the reaction was extracted with AcOEt 3 times.
The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated by rotavapor. The residue was purified by FC on RP using basic conditions (eluting from 5:95 of CH3CN/H2O + 0.1 % of NH4OH to 40:60 of CH3CN/H2O + 0.1 % of NH4OH) affording the product of formula (2-cyclopropyl-6-pyrrolidin-l-yl-3H-benzimidazol-5-yl)-(4-methyl-2- phenylpiperazin-l-yl)methanone.
Yield: 45 mg
NMR (400 MHz, DMSO-d6) δ 11.95 (s, 1H), 7.76 -7.45 (m, 2H), 7.37 (dt, 2H), 7.28 (t, 1H), 7.21 -
7.14 (m, 1H), 7.12 -6.69 (m, 1H), 5.92 -3.44 (m, 2H), 3.17 -2.53 (m, 6H), 2.46 -2.12 (m, 5H), 2.05 -
1.78 (m, 5H), 1.73 -1.60 (m, 1H), 1.10 - 0.83 (m, 4H); LC-MS: m/z 430.52 (MH+).
Preparation: (4-methyl-2-phenylpiperazin-l-yl)-(2-methyl-6-pyrrolidin-l-yl-3H-benzimidazol-5- yl)methanone
Figure imgf000244_0002
Example 114 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 113 (2-cyclopropyl-6-pyrrolidin-l-yl-3H-benzimidazol-5-yl)-(4-methyl-2-phenylpiperazin- l-yl)methanone wherein acetaldehyde was used instead of cyclopropanecarboxaldehyde. The title compound was obtained in a yield of 49 % (47 mg).
NMR (400 MHz, DMSO-d6) δ 12.05 -11.81 (m, 1H), 7.78 -7.44 (m, 1H), 7.44 -7.21 (m, 3H), 7.20 -
6.93 (m, 2H), 6.85 -6.67 (m, 1H), 5.88 -3.44 (m, 2H), 3.24 -2.74 (m, 6H), 2.64 -2.56 (m, 1H), 2.45 -
2.11 (m, 5H), 2.07 -1.87 (m, 5H), 1.80 -1.59 (m, 2H); LC-MS: m/z 403.68 (MH+).
Scheme 24
Figure imgf000245_0001
Step 1
Compound of formula II may be obtained via acyl chloride formation from compound of formula I
(commercially available from Enamine) by treatment with a suitable chlorinating agent, such as
SOCI2, typically at 90 °C, followed by coupling with l-Methyl-3-phenylpiperazine (commercially available from Enamine) in the presence of a suitable organic base, e.g. TEA. The reaction is carried out in a suitable solvent such as DCM, typically at room temperature. The reaction takes about 16 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by Buchwald reaction with the proper amide, e.g. cyclopropanecarboxamide, in the presence of a suitable palladium precatalyst, e.g. XPhos Pd G3, and a suitable base, such as Cs2CO3. The reaction is carried out in a suitable solvent, e.g. 1, 4-dioxane, typically at 80 °C. The reaction takes about 12 hours to complete. Step 3
Compound of formula IV may be obtained from compound of formula III by Buchwald reaction with the proper amine, e.g. pyrrolidine, in the presence of a suitable palladium precatalyst, e.g. XPhos
Pd G3, and a suitable base, such as K2CO3. The reaction is carried out in a suitable solvent, e.g. 1, 4- dioxane, typically at 100 °C. The reaction takes about 12 hours to complete.
Scheme 24, Cpd (II) Preparation: (2,6-dichloro-5-fluoropyridin-3-yl)-(4-methyl-2-phenylpiperazin-l- yl)methanone
Figure imgf000246_0001
A mixture of 2,6-dichloro-5-fluoropyridine-3-carboxylic acid (1.0 g, 4.76 mmol) and thionyl dichloride (3.47 mL, 47.62 mmol) was heated to reflux for 3 h. After this time the reaction was cooled down to RT and concentrated under vacuum. The residue was dissolved in DCM (1.667 mL) and cooled to 0 °C. After 10 min a solution of DIPEA (1.66 mL, 9.52 mmol) and l-methyl-3- phenylpiperazine (0.84 g, 4.76 mmol) in DCM (10 mL) was added and the reaction was stirred at RT overnight. After this time the reaction was diluted with DCM and washed with a s.s. of NaHCO33 times. The organic phase was washed with brine, dried over a phase separator and concentrated under vacuum affording the product of formula (2,6-dichloro-5-fluoropyridin-3-yl)-(4-methyl-2- phenylpiperazin-l-yl)methanone. The product was used in the next step without further purification.
Yield: 1.6 g
1H NMR (400 MHz, DMSO-d6) δ 8.42 -8.10 (m, 1H), 7.53 (dd, 1H), 7.44 -7.19 (m, 4H), 5.83 -3.45 (m,
1H), 3.16 -2.56 (m, 2H), 2.47 -2.29 (m, 1H), 2.25 -2.12 (m, 3H), 2.12 -1.89 (m, 1H), 0.95 (d, 2H); LC-
MS: m/z 368.04 (MH+)
Scheme 24. Cpd (III) Preparation: N-[6-chloro-3-fluoro-5-(4-methyl-2-phenylpiperazine-l- carbonyl)pyridin-2-yl]cyclopropanecarboxamide
Figure imgf000247_0001
A solution of (2,6-dichloro-5-fluoropyridin-3-yl)-(4-methyl-2-phenylpiperazin-l-yl)methanone
(200.0 mg, 0.540 mmol), cyclopropanecarboxamide (50.84 mg, 0.600 mmol), dicesium carbonate
(356.13 mg, 1.09 mmol) and XPHOS PD G3 (23.01 mg, 0.030 mmol) in dry 1,4-Dioxane (4.964 mL) was degassed by Schlenk technique. The resulting mixture was heated to 80 °C and stirred ON. The day after the reaction mixture was diluted with EtOAc and filtered through a pad of celite. The filtrate was concentrated under reduced pressure and purified by FC on RP using basic conditions
(eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 60:40 of CH3CN/H2O + 0.1% of HCOOH). Pure fraction were collected and evaporated under vacuum affording the product of formula (N-[6- chloro-3-fluoro-5-(4-methyl-2-phenylpiperazine-l-carbonyl)pyridin-2- yl]cyclopropanecarboxamide.
Yield: 65 mg
1H NMR (500 MHz, DMSO-d6 ) δ ppm 11.28 - 10.70 (m, 1 H), 8.33 - 7.83 (m, 1 H), 7.60 - 7.31 (m, 4
H), 7.31 - 7.24 (m, 1 H), 5.85 - 4.58 (m, 1 H), 3.57 - 3.24 (m, 1 H), 4.46 - 3.19 (m, 1 H), 3.17 - 2.84 (m,
1 H), 2.83 - 2.54 (m, 1 H), 2.50 (dt, 1 H), 2.26 - 2.14 (m, 3 H), 2.13 - 1.97 (m, 1 H), 1.97 - 1.83 (m, 1
H), 0.96 - 0.70 (m, 4 H); LC-MS: m/z 417.17 (MH+).
Scheme 24, Cpd (IV) Preparation: N-[3-fluoro-5-(4-methyl-2-phenylpiperazine-l-carbonyl)-6- pyrrolidin-l-ylpyridin-2-yl]cyclopropanecarboxamide
Figure imgf000247_0002
Example 115
A solution of N-[6-chloro-3-fluoro-5-(4-methyl-2-phenylpiperazine-l-carbonyl)pyridin-2- yl]cyclopropanecarboxamide (65.0 mg, 0.160 mmol), potassium carbonate (0.04 mL, 0.310 mmol), XPHOS PD G3 (65.99 mg, 0.080 mmol) and pyrrolidine (11.09 mg, 0.160 mmol) in 1,4- Dioxane (1.5 mL) was by Schlenk technique and stirred ON at 100 °C. The day after the reaction was cooled down to RT, diluted with water and extracted with EtOAc 3 times. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of cHex to cHex/AcOEt 50:50) and again by FC on
RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O
+ 0.1% of HCOOH). Pure fractions were collected and concentrated under vacuum. The residue was taken up with a s.s. of NaHCO3 and extracted 3 times with AcOEt. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula N-[3-fluoro-5-(4-methyl-2-phenylpiperazine-l-carbonyl)-6-pyrrolidin-l- ylpyridin-2-yl]cyclopropanecarboxamide.
Yield: 6 mg
1H NMR (500 MHz, DMSO-d6 ) δ ppm 10.39 - 10.07 (m, 1 H), 7.83 - 7.00 (m, 6 H), 5.84 - 4.74 (m, 1
H), 4.56 - 2.60 (m, 8 H), 2.47 - 1.80 (m, 5 H), 2.03 - 1.47 (m, 5 H), 0.91 - 0.63 (m, 4 H); LC-MS: m/z
452.23 (MH+).
Scheme 25
Figure imgf000248_0001
Step 1
Compound of formula II may be obtained by Ullmann reaction between compound of formula I
(synthesized according to the Scheme 3) and the appropriate commercially available pyrazole in the presence of a suitable catalyst, e.g. Cul, a suitable ligand, e.g. trans-N,N'-Dimethylcyclohexane-l,2- diamine, typically at 120 °C. The reaction takes about 12 hours to complete.
Preparation: N-[3-[3-(difluoromethyl)pyrazol-l-yl]-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000249_0002
Example 116
To a mixture of N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide (100.0 mg, 0.240 mmol), tripotassium phosphate
(107.03 mg, 0.500 mmol) and 3-(difluoromethyl)-lH-pyrazole (33.55 mg, 0.280 mmol) copper (I) iodide (2.27 mg, 0.010 mmol) and N,N'-Dimethylcyclohexane-l,2-diamine (3.37 mg, 0.020 mmol) were added. The reaction was degassed with 5 cycles of N2/vacuum and stirred ON at 120 °C. The day after the reaction mixture was cooled down to RT, diluted with EtOAc and filtered through a pad of celite. The organic layer was evaporated under reduced pressure and purified by FC on RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 40:60 of CH3CN/H2O +
0.1% of HCOOH) affording the product of formula N-[3-[3-(difluoromethyl)pyrazol-l-yl]-4-(l,l- dioxo-l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide.
Yield: 3.4 mg
1H NMR (400 MHz, MeOD) δ 8.11 (d, 1H), 8.05 (d, 1H), 7.61 (dd, 1H), 7.45 (d, 1H), 7.01 -6.67 (m,
2H), 4.49 (d, 1H), 3.97 (d, 1H), 3.78 (dd, 2H), 3.45 (d, 2H), 3.25 -3.14 (m, 2H), 3.00 (d, 1H), 1.79 (tt,
1H), 1.10 -0.86 (m, 4H); LC-MS: 439.11 (MH+).
Preparation: N-[3-(3-cyanopyrazol-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000249_0001
Example 117 The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 116 N-[3-[3-(difluoromethyl)pyrazol-l-yl]-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide wherein lH-pyrazole-3-carbonitrile was used instead of 3-(difluoromethyl)-lH-pyrazole. The title compound was obtained in a yield of 4 % (6 mg).
1H NMR (400 MHz, DMSO-d6 ) δ ppm 10.68 (s, 1 H), 8.41 (d, 1 H), 8.05 (d, 1 H), 7.67 (dd, 1 H), 7.55
(d, 1 H), 7.24 (d, 1 H), 4.05 - 3.51 (m, 4 H), 3.43 - 2.90 (m, 4 H), 1.88 - 1.70 (m, 1 H), 0.92 - 0.72 (m, 4
H); LC-MS: m/z 414.18 (MH+).
Preparation: N-[3-(3-tert-butylpyrazol-l-yl)-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide
Figure imgf000250_0001
Example 118
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 116 N-[3-[3-(difluoromethyl)pyrazol-l-yl]-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide wherein 3-tert-butyl-lH-pyrazole was used instead of
3-(difluoromethyl)-lH-pyrazole. The title compound was obtained in a yield of 15 % (9 mg).
1H NMR (400 MHz, DMSO) δ 10.53 (s, 1H), 7.88 (dd, 2H), 7.59 (dd, 1H), 7.44 (d, 1H), 6.42 (d, 1H),
4.13 (s, 1H), 3.75 (t, 1H), 3.61 (dd, 1H), 3.36 (s, 1H), 3.19 -3.09 (m, 1H), 2.99 (dd, 1H), 2.52 (d, 2H),
1.80 (p, 1H), 1.26 (s, 9H), 0.83 (d, 4H); LC-MS: m/z 445.16 (MH+).
Preparation: N-[4-(4,4-difluoropiperidine-l-carbonyl)-3-[3-(trifluoromethyl)pyrazol-l- yl)phenyl)cyclopropanecarboxamide
Figure imgf000251_0001
Example 119
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 116 N-[3-[3-(difluoromethyl)pyrazol-l-yl]-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide wherein N-[3-bromo-4-(4,4-difluoropiperidine-l- carbonyl)phenyl]cyclopropanecarboxamide and 3-(trifluoromethyl)pyrazole were used instead of N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)phenyl]cyclopropanecarboxamide and 3-
(difluoromethyl)-lH-pyrazole respectively. The title compound was obtained in a yield of 44 % (60 mg).
1H NMR (400 MHz, DMSO) δ 10.63 (s, 1H), 8.33 (dd, 1H), 8.01 (d, 1H), 7.66 (dd, 1H), 7.44 (d, 1H),
7.00 (d, 1H), 3.98 - 3.82 (m, 1H), 3.38 - 3.31 (m, 3H), 2.17 - 1.63 (m, 5H), 0.89 - 0.80 (d, 4H); LC-MS: m/z 443.14 (MH+).
Scheme 26
Figure imgf000252_0001
Step 1
Compound of formula II may be obtained via Suzuki coupling between compound of formula I and the appropriate heterocycle boronic ester, e.g. l-(propan-2-yl)-3-(tetramethyl-l,3,2-dioxaborolan-
2-yl)-lH-pyrazole (commercially available from Enamine), in the presence of a suitable precatalyst, e.g. XPhos Pd G2, a suitable base, e.g. K3PO4, in a suitable mixture of solvents, e.g. 1,2- dimethoxyethane / water, typically at 90 °C. The reaction takes about 3 hours to complete
Step 2
Compound of formula III may be obtained by nitrile cyclization of compound of formula II with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about 6 hours to complete.
Step 3
Compound of formula IV may be obtained from compound of formula III by hydrolysis with LiOH, in a suitable mixture of solvents such as MeOH/THF/H2O, typically at 50 °C. The reaction takes about
12 hours to complete. Step 4
Compound of formula V may be obtained from compound of formula IV by coupling with the appropriate amine, e.g. 4-piperidinecarbonitrile, in the presence of a suitable coupling agent, e.g.
HATU, and an organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent such as DMF, typically at room temperature. The reaction takes about 12 hours to complete.
Scheme 26, Cpd (II) Preparation: methyl 4-cyano-2-(l-isopropylpyrazol-3-yl)benzoate
Figure imgf000253_0001
A mixture of X-phos aminobiphenyl palladium chloride precatalyst (98.2 mg, 0.120 mmol), tripotassium phosphate (795.85 mg, 3.75 mmol), l-(propan-2-yl)-3-(tetramethyl-l, 3,2- dioxaborolan-2-yl)-lH-pyrazole (236.07 mg, 1 mmol) and methyl 2-bromo-4-cyanobenzoate (300.0 mg, 1.25 mmol) in Water (0.397 mL)/l,2-dimethoxyethane (7.935 mL) was degassed with Shlenk line technique and then stirred at 90 °C for 3h. After this time the reaction was cooled down.
Water was added to the mixture and aqueous phase was extracted with AcOEt 3 times. Organic phases were combined, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of cHex to 40:60 of cHex/ AcOEt) affording the product of formula methyl 4-cyano-2-(l-isopropylpyrazol-3-yl)benzoate.
Yield: 250 mg
1H NMR (400 MHz, DMSO-d6) δ 8.00 -7.96 (m, 2H), 7.93 -7.89 (m, 1H), 7.86 (d, 1H), 6.67 (d, 1H),
4.52 (p, 1H), 3.76 (s, 3H), 1.42 (d, 6H); LC-MS: m/z 270.24 (MH+).
Scheme 26, Cpd (III) Preparation: methyl 2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-
3-yl)benzoate
Figure imgf000254_0001
A mixture of methyl 4-cyano-2-(l-isopropylpyrazol-3-yl)benzoate (250.0 mg, 0.930 mmol), dicesium carbonate (907.41 mg, 2.78 mmol), ethanimidamide hydrochloride (131.65 mg, 1.39 mmol) and CuBr (6.66 mg, 0.050 mmol) in DMSO (10 mL) was stirred at 120 °C for 6 h. After this time the reaction was cooled down to RT and H2O was added. The mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of
DCM to 95:5 of DCM/MeOH) affording the product of formula methyl 2-(l-isopropylpyrazol-3-yl)-
4-(5-methyl-4H-l,2,4-triazol-3-yl)benzoate.
Yield: 118 mg
1H NMR (400 MHz, DMSO-d6) δ 13.81 (s, 1H), 8.16 -7.99 (m, 2H), 7.88 -7.63 (m, 2H), 6.52 (d, 1H),
4.51 (p, 1H), 3.74 (s, 3H), 2.40 (s, 3H), 1.43 (d, 6H); LC-MS: m/z 326.4 (MH+).
Scheme 26, Cpd (IV) Preparation: 2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)benzoic acid
Figure imgf000254_0002
To a solution of methyl 2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)benzoate
(118.0 mg, 0.360 mmol) in Methanol (0.400 mL)/THF (2 mL) a solution of lithium hydroxide (13.03 mg, 0.540 mmol) in Water (0.400 mL) was added. The reaction was stirred at 50 °C ON. The day after the reaction was cooled down to RT, acidified with HCI 3 N and concentrated under vacuum. The residue was purified by FC on reverse phase using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 50:50 of CH3CN/H2O + 0.1% of HCOOH) affording the product of formula 2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)benzoic acid.
Yield: 80 mg
1H NMR (400 MHz, DMSO-d6) δ 13.74 (bs, 1H), 12.89 (bs, 1H), 8.15 -7.99 (m, 2H), 7.85 -7.63 (m,
2H), 6.48 (s, 1H), 4.52 (p, 1H), 2.43 (s, 3H), 1.46 (d, 6H); LC-MS: m/z 312.35 (MH+).
Scheme 26, Cpd (V) Preparation: l-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)benzoyl]piperidine-4-carbonitrile
Figure imgf000255_0001
Example 120
To a solution of , 2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)benzoic acid (33.3 mg, 0.110 mmol) in DMF (0.491 mL), N,N-Diisopropylethylamine (0.06 mL, 0.320 mmol)and HATU
(52.87 mg, 0.140 mmol) were added. The reaction was stirred at RT for 15 min. After this time 4- piperidinecarbonitrile (11.78 mg, 0.110 mmol) was added and the reaction was stirred ON at
RT. After this time the reaction was diluted with a saturated aqueous solution of NaHCO3 and extracted with AcOEt 3 times. The combined organic fractions were dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of DCM to EtOAc 100%) and again by FC on RP using acid conditions (eluting from 5:95 of CH3CN/H2O + 0.1% of HCOOH to 20:80 of CH3CN/H2O + 0.1% of HCOOH). Pure fractions were collected and evaporated.
The residue was taken up with a s.s. of NaHCO3 and extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum affording the product of formula l-[2-(l-isopropylpyrazol-3-yl)-4-(5-methyl-4H-l,2,4- triazol-3-yl)benzoyl]piperidine-4-carbonitrile.
Yield: 19 mg.
1H NMR (400 MHz, DMSO-d6) δ ppm 8.02 (dt, 1 H), 7.77 - 7.87 (m, 3 H), 6.49 (dd, 1 H), 4.53 (e, 1 H),
3.88 - 4.10 (m, 1 H), 3.35 - 3.44 (m, 1 H), 3.00 - 3.11 (m, 2 H), 2.89 (dt, 1 H), 2.37 - 2.43 (m, 3 H), 1.87
- 1.98 (m, 1 H), 1.51 - 1.82 (m, 3 H), 1.41 - 1.50 (m, 6 H), 0.98 - 1.10 (m, 1 H); LC-MS: m/z 404.37
(MH+). Scheme 27
Figure imgf000256_0002
Step 1
Compound of formula II may be obtained by Ullmann reaction between compound of formula I
(synthesized according to the Scheme 4) and the appropriate commercially available pyrazole in the presence of a suitable catalyst, e.g. Cul, a suitable ligand, e.g. trans-N,N'-Dimethylcyclohexane-l,2- diamine, typically at 120 °C. The reaction takes about 2 days to complete.
Step 2
Compound of formula III may be obtained by nitrile cyclization of compound of formula II with ethanimidamide hydrochloride in presence of a suitable catalyst, e.g. CuBr, a suitable base, e.g.
Cs2CO3, in a suitable solvent, such as DMSO, typically at 120 °C. The reaction takes about 2 hours to complete.
Scheme 27, Cpd (ll)*a Preparation: 3-(3-cyclopropylpyrazol-l-yl)-4-[4-(trifluoromethyl)piperidine- l-carbonyl]benzonitrile
Figure imgf000256_0001
In a via 3-bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile (200.0 mg, 0.550 mmol), tripotassium phosphate (250.36 mg, 1.16 mmol), 3-cyclopropyl-lH-pyrazole (71.86 mg,
0.660 mmol) were suspended in dry 1,4-Dioxane (3.038 mL). The resulting suspension was degassed bubbling N2 for 15 minutes, then copper (I) iodide (5.3 mg, 0.030 mmol) and N,N'-
Dimethylcyclohexane-l,2-diamine (7.88 mg, 0.060 mmol) were added under nitrogen flux and degassed again with Schlenk line technique. The vial was sealed and stirred at 120 °C for 2 days. The day after the reaction mixture was diluted with EtOAc and filtered through a pad of celite. The organic layer was washed with brine, dried over Na2SO4, filtered and evaporated under reduced pressure affording the product of formula 3-(3-cyclopropylpyrazol-l-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile, which was used in the next step without further purification.
Yield: 180 mg
LC-MS: m/z 389.20 (MH+).
Scheme 27, Cpd (ll)*b Preparation: 3-(3-tert-butylpyrazol-l-yl)-4-(4,4-difluoropiperidine-l- carbonyl)benzonitrile
Figure imgf000257_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound 3-
(3-cyclopropylpyrazol-l-yl)-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile wherein bromo-4-(4,4-difluoropiperidine-l-carbonyl)benzonitrile and 3-tert-butyl-lH-pyrazole were used instead of 3-bromo-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile and 3-cyclopropyl-lH- pyrazole respectively. The title compound was obtained in a quantitative yield (150 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 8.20 (s, 2H), 7.90 - 7.83 (m, 1H), 7.70 - 7.63 (m, 1H), 6.51 -
6.45 (m, 1H), 4.02 - 3.94 (m, 1H), 3.42 - 3.32 (m, 1H), 3.08 - 2.95 (m, 1H), 2.20 - 1.85 (m, 3H), 1.72 -
1.55 (m, H), 1.25 (s, 9H); LC-MS: m/z 373.36 (MH+).
Scheme 27, Cpd (lll)*a Preparation: [2-(3-cyclopropylpyrazol-l-yl)-4-(5-methyl-4H-l,2,4-triazol-3- yl)phenyl]-[4-(trifluoromethyl)piperidin-l-yl]methanone
Figure imgf000257_0002
Example 121
A mixture of 3-(3-cyclopropylpyrazol-l-yl)-4-[4-(trifluoromethyl)piperidine-l-carbonyl]benzonitrile
(180.0 mg, 0.460 mmol), dicesium carbonate (453.01 mg, 1.39 mmol), ethanimidamide hydrochloride (65.72 mg, 0.700 mmol) and CuBr (3.32 mg, 0.020 mmol) in DMSO (5.143 mL) was stirred at 120 °C for 2h. After this time the reaction was cooled down to RT and H2O was added. The mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of DCM to DCM/MeOH 9:1) affording the product of formula [2-
(3-cyclopropylpyrazol-l-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-(trifluoromethyl)piperidin- l-yl]methanone.
Yield: 16 mg
1H NMR (400 MHz, DMSO-d6) δ 13.86 (bs, 1H), 8.12 -8.09 (m, 1H), 8.06 -7.87 (m, 2H), 7.51 -7.39 (m,
1H), 6.30 -6.15 (m, 1H), 4.66 -4.44 (m, 1H), 3.47 -3.36 (m, 1H), 3.04 -2.91 (m, 1H), 2.78 -2.57 (m, 2H),
2.41 (s, 3H), 2.00 -1.62 (m, 3H), 1.54 -1.06 (m, 2H), 0.97 -0.84 (m, 2H), 0.79 -0.56 (m, 2H); LC-MS: m/z 445.2 (MH+).
Preparation: [2-(3-tert-butylpyrazol-l-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4- difluoropiperidin-l-yl)methanone
Figure imgf000258_0001
Example 122
The synthesis of the title compound was effected analogously to the synthesis of the compound of
Example 121 [2-(3-cyclopropylpyrazol-l-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-[4-
(trifluoromethyl)piperidin-l-yl]methanone wherein 3-(3-tert-butyl pyrazol-l-yl )-4-(4,4- difluoropiperidine-l-carbonyl)benzonitrile was used instead of 3-(3-cyclopropylpyrazol-l-yl)-4-[4-
(trifluoromethyl)piperidine-l-carbonyl]benzonitrile. The title compound was obtained in a yield of
35% (50 mg).
NMR (400 MHz, DMSO-d6) δ 8.08 (d, 1H), 7.99 - 7.89 (m, 2H), 7.41 (d, 1H), 6.43 (d, 1H), 3.66 (q,
2H), 3.14 - 3.02 (m, 1H), 2.30 (s, 3H), 2.12 - 1.99 (m, 1H), 1.94 - 1.77 (m, 2H), 1.43 - 1.21 (m, 11H);
LC-MS: m/z 429.26 (MH+). Scheme 28
Figure imgf000259_0001
Step 1
Compound of formula II may be obtained by reaction between compound I (synthesized according to the Scheme 3) and 2-pyrrolidinone in the presence of a suitable catalyst, e.g. Cul, a suitable ligand, e.g. trans-N,N'-Dimethylcyclohexane-l,2-diamine, and a suitable base, e.g. K2CO3. The reaction is carried out in a suitable solvent, such as 1, 4-dioxane, typically at 120 °C. The reaction takes about 12 hours to complete. Alternatively compound of formula II may be obtained by
Buchwald reaction between compound I (synthesized according to the Scheme 3) and 2- pyrrolidinone in the presence of a suitable catalyst, e.g. Pd2(dba)s, a suitable ligand, such as XPhos, and a suitable base, e.g. Cs2CO3. The reaction is carried out in a suitable solvent, such as 1, 4- dioxane, typically at 110 °C. The reaction takes about 12 hours to complete.
Preparation: N-[4-(l,l-dioxo-l,4-thiazinane-4-carbonyl)-3-(2-oxopyrrolidin-l- yl)phenyl]cyclopropanecarboxamide
Figure imgf000259_0002
Example 123
To a suspension of N-[3-bromo-4-(l,l-dioxo-l,4-thiazinane-4- carbonyl)phenyl]cyclopropanecarboxamide (200.0 mg, 0.500 mmol), potassium carbonate (137.77 mg, 1 mmol) and 2-pyrrolidinone (0.05 mL, 0.600 mmol) in 1,4-Dioxane (3 mL) N,N'-
Dimethylcyclohexane-l,2-diamine (0.02 mL, 0.100 mmol) and copper (I) iodide (9.49 mg, 0.050 mmol) were added. The mixture was degassed with N2 for 10 min and then heated to 120 °C ON.
The day after the reaction was diluted with AcOEt and washed with brine. The organic phase was dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on silica gel (eluting from 100% of DCM to DCM/MeOH 9:1) affording the product of formula N-[4-(l,l- dioxo-l,4-thiazinane-4-carbonyl)-3-(2-oxopyrrolidin-l-yl)phenyl]cyclopropanecarboxamide.
Yield: 54 mg
1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 7.67 (d, 1H), 7.47 (dd, 1H), 7.38 (d, 1H), 4.21 - 3.56
(m, 6H), 3.39 (s, 1H), 3.21 (t, 3H), 2.45 - 2.31 (m, 1H), 2.19 - 1.63 (m, 4H), 0.83 (d, 4H); LC-MS: m/z
406.2 (MH+)
Preparation: N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(2-oxopyrrolidin-l- yl)phenyl]cyclopropanecarboxamide
Figure imgf000260_0001
Example 124
To a suspension of N-[3-bromo-4-(4-methyl-2-phenylpiperazine-l- carbonyl)phenyl]cyclopropanecarboxamide (200.0 mg, 0.450 mmol), 2-pyrrolidinone (57.71 mg,
0.680 mmol) and (5-diphenylphosphino-9,9-dimethyl-4-xanthenyl)-diphenylphosphine (130.81 mg,
0.230 mmol) in 1,4-Dioxane (1.5 mL) dicesium carbonate (294.63 mg, 0.900 mmol) and (1E,4E)-1,5- diphenyl-3-penta-l,4-dienone palladium (62.1 mg, 0.070 mmol) were added. The mixture was degassed with N2 for 10 min and then heated to 110 °C ON. The day after the reaction was diluted with AcOEt and washed with brine. The organic phase was dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from cHex/ AcOEt 80:20 to cHex/AcOEt 40:60) and again by FC on RP using basic conditions (eluting from
5:95 of CH3CN/H2O + 0.1 % of NH4OH to 30:70 of CH3CN/H2O + 0.1 % of NH4OH) affording the product of formula (N-[4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(2-oxopyrrolidin-l- yl)phenyl]cyclopropanecarboxamide.
Yield: 66 mg
1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 7.73 - 7.62 (m, 1H), 7.59 - 7.40 (m, 2H), 7.41 - 7.21 (m,
5H), 5.86 - 4.34 (m, 1H), 3.82 - 3.35 (m, 4H), 2.89 (d, 2H), 2.39 - 1.67 (m, 10H), 0.86 - 0.79 (m, 4H);
LC-MS: m/z 447.25 (MH+). Scheme 29
Figure imgf000261_0002
Step 1
Compound of formula II may be obtained from the commercially available SnAP reagent (Compound
I) by reaction with the appropriate aromatic aldehyde, followed by Cu mediated cyclization. The reaction is carried out in a proper solvent, e.g. DCM, typically at RT. The reaction takes about 12 hours to complete.
Scheme 29, Cpd (ll)*a Preparation: tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate
Figure imgf000261_0001
To a solution of SnAP Pip reagent (771.19 mg, 1.66 mmol) in dry DCM (8.3 mL) at room temperature
3-methylbenzaldehyde (200.0 mg, 1.66 mmol) and previously activated MS 3A were added. The reaction was stirred at rt overnight and then filtered through a short layer of celite, washing with
DCM. The filtrate was concentrated under reduced pressure to afford the imine. Separately, 2, 6-
Lutidine (178.36 mg, 1.66 mmol) was added in one portion to a suspension of dry Copper(ll) trifluoromethanesulfonate (602.05 mg, 1.66 mmol) in anhydrous HFIP (6.6 mL, 1.66 mmol) and stirred at RT for lh, during which time a homogeneous suspension was formed. A solution of imine in dry DCM (26 mL) was added in one portion and the resulting mixture was stirred at rt overnight. After 18 h, the reaction was quenched at rt with 15 mL of a 10% aqueous solution of
NH40H and stirred vigorously for 15 min. The layers were separated and the aqueous layer was extracted with DCM x3. The combined organic layers were washed with H2O, brine, dried using a phase separator and concentrated. The crude was purified by FC on RP using basic conditions
(eluting from 5:95 of CH3CN/H2O + 0.1 % of NH4OH to 30:70 of CH3CN/H2O + 0.1 % of NH4OH) affording the product of formula tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate.
Yield: 336 mg
1H NMR (400 MHz, DMSO-d6) δ ppm 7.25 - 7.15 (m, 3H), 7.08 (d, 1H), 3.92 - 3.74 (m, 2H), 3.51 (dd,
1 H), 2.99 - 2.57 (m, 5H), 2.29 (s, 3H), 1.40 (s, 9H); LC-MS: m/z 277.46 (MH+).
Scheme 29, Cpd (ll)*b Preparation: tert-butyl 3-(3-chloro-5-methylphenyl)piperazine-l-carboxylate
Figure imgf000262_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate wherein 3-chloro-5-methylbenzaldehyde was used instead of 3-methylbenzaldehyde. The title compound was obtained in a yield of 69 %
(276 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.25 (s, 1H), 7.22 - 7.14 (m, 2H), 3.88 - 3.74 (m, 2H), 3.54 (dt,
1H), 2.97 - 2.88 (m, 1H), 2.87 - 2.76 (m, 2H), 2.69 - 2.55 (m, 2H), 2.30 (s, 3H), 1.40 (s, 9H); LC-MS: m/z 311.14 (MH+).
Scheme 29, Cpd (ll)*c Preparation: tert-butyl 3-(4-methylphenyl)piperazine-l-carboxylate
Figure imgf000262_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate wherein 4-methylbenzaldehyde was used instead of 3-methylbenzaldehyde. The title compound was obtained in a yield of 66 % (306 mg). 1H NMR (400 MHz, DMS0-d6) δ ppm 7.32 - 7.10 (m, 4H), 3.92 - 3.74 (m, 2H), 3.56 - 3.47 (m, 1H),
2.99 - 2.58 (m, 5 H), 2.28 (s, 3H), 1.40 (s, 9H); LC-MS: m/z 277.71 (MH+).
Scheme 29, Cpd (ll)*d Preparation: tert-butyl 3-[2-(trifluoromethyl)phenyl]piperazine-l- carboxylate
Figure imgf000263_0001
The synthesis of the title compound was effected analogously to the synthesis of the compound tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate wherein 2-(trifluoromethyl)benzaldehyde was used instead of 3-methylbenzaldehyde. The title compound was obtained in a yield of 70 %
(200 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.96 (d, 1H), 7.76 - 7.64 (m, 2H), 7.49 (t, 1H), 3.96 - 3.81 (m,
3H), 2.96 (d, 2H), 2.89 - 2.74 (m, 1H), 2.74 - 2.59 (m, 2H), 1.38 (s, 9H); LC-MS: m/z 275.12 (MH+).
Scheme 29, Cpd (ll)*e Preparation: tert-butyl 3-(3-cyanophenyl)piperazine-l-carboxylate
Figure imgf000263_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate wherein 3-formylbenzonitrile was used instead of 3-methylbenzaldehyde. The title compound was obtained in a yield of 51 % (168 mg).
1H NMR (400 MHz, DMSO-d6) δ ppm 7.85 (s, 1 H), 7.81 - 7.71 (m, 2 H), 7.59 - 7.53 (m, 1 H), 3.92 -
3.75 (m, 2 H), 3.70 - 3.61 (m, 1 H), 2.98 - 2.78 (m, 3 H), 2.70 - 2.57 (m, 2 H), 1.40 (s, 9 H); LC-MS: m/z
288.23 (MH+).
Scheme 29, Cpd (ll)*f Preparation: tert-butyl 3-(4,5-dimethyl-l,3-thiazol-2-yl)piperazine-l- carboxylate
Figure imgf000264_0002
The synthesis of the title compound was effected analogously to the synthesis of the compound tert-butyl 3-(3-methylphenyl)piperazine-l-carboxylate wherein 4,5-dimethylthiazole-2- carboxaldehyde was used instead of 3-methylbenzaldehyde. The title compound was obtained in a yield of 64 % (203 mg).
1H NMR (400 MHz, DMSO-d6) δ 3.99 (s, 1H), 3.86 -3.75 (m, 1H), 3.71 (d, 1H), 3.10 -3.05 (m, 1H), 2.98
-2.80 (m, 2H), 2.71 -2.62 (m, 2H), 2.30 (s, 3H), 2.23 (s, 3H), 1.41 (s, 9H); LC-MS: m/z 297.61 (MH+).
Scheme 30
Figure imgf000264_0001
Step 1
Compound of formula II may be obtained by coupling reaction between compound of formula I
(commercially available from Zantec) and the appropriate amine, e.g. l-methyl-3-phenylpiperazine, in presence of a suitable coupling agent, e.g. HATU, a suitable organic base, e.g. DIPEA. The reaction is carried out in a suitable solvent, e.g. DMF, typically at RT. The reaction takes about 2 hours to complete.
Step 2
Compound of formula III may be obtained from compound of formula II by SNAr substitution with
3-(propan-2-yl)pyrrolidine in a suitable aprotic solvent, e.g. DMSO, typically at 120° C. The reaction takes about 2 days to complete.
Scheme 30, Cpd (II) Preparation: 3-fluoro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)benzonitrile
Figure imgf000265_0002
A solution of 4-cyano-2-fluorobenzoic acid (500.0 mg, 3.03 mmol), N,N-Diisopropylethylamine (2.64 mL, 15.14 mmol) and HATU (1727.06 mg, 4.54 mmol) in DMF (15.14 mL) was stirred for 15 minutes at RT. Then l-methyl-3-phenylpiperazine (640.48 mg, 3.63 mmol) was added and the reaction was stirred at RT for 2 hours. A saturated solution of NaHCO3 was added to the reaction mixture and the aqueous phase was extracted with EtOAc (x3). The organic portions were collected, washed with brine, residual water was removed by Na2SO4, the solvent was filtered and evaporated under reduced pressure. The crude was purified by FC on silica gel (eluting from 100% of cHex to cHex/AcOEt 1:1) affording the product of formula 3-fluoro-4-(4-methyl-2-phenylpiperazine-l- carbonyl)benzonitrile.
Yield: 880 mg
NMR (400 MHz, DMSO-d6) δ 8.03 (d, 1H), 7.89 - 7.63 (m, 2H), 7.54 - 7.22 (m, 5H), 5.91 - 4.26 (m,
1H), 3.59 - 3.43 (m, 1H), 3.24 - 2.60 (m, 3H), 2.41 - 2.28 (m, 1H), 2.20 (s, 3H), 2.07 - 1.82 (m, 1 H); LC-
MS: m/z 324.20 (MH+).
Scheme 30, Cpd (III) Preparation: 4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(3-propan-2- ylpyrrolidin-l-yl)benzonitrile
Figure imgf000265_0001
To a solution of 3-fluoro-4-(4-methyl-2-phenylpiperazine-l-carbonyl)benzonitrile (300.0 mg, 0.930 mmol) and 3-(propan-2-yl)pyrrolidine hydrochloride (166.62 mg, 1.11 mmol) in DMSO (8.737 mL)
N,N-Diisopropylethylamine (0.4 mL, 2.32 mmol) was added. The mixture was stirred at 120 °C for 2 days. After this time the reaction was cooled down to RT and diluted with H2O and was extracted with Acoet. The combined organic fractions were washed with brine, dried over Na2SO4 and concentrated under vacuum. The crude was purified by FC on NH column(eluting from 100% of cHex to cHex/AcOEt 75:25) affording the product of formula 4-(4-methyl-2-phenylpiperazine-l- carbonyl)-3-(3-propan-2-ylpyrrolidin-l-yl)benzonitrile as mixture of diasteroisomers.
Yield: 95 mg
LC-MS: m/z: m/z 471.34 (MH+).
Scheme 30, Cpd (IV) Preparation: (4-methyl-2-phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-
3-yl)-2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone
Figure imgf000266_0001
Example 125a, Example 125b, Example 125c, and Example 125d
A mixture of 4-(4-methyl-2-phenylpiperazine-l-carbonyl)-3-(3-propan-2-ylpyrrolidin-l- yl)benzonitrile (95.0 mg, 0.230 mmol), dicesium carbonate (222.92 mg, 0.680 mmol), ethanimidamide hydrochloride (32.34 mg, 0.340 mmol) and bromocopper (1.64 mg, 0.010 mmol) in DMSO (3.5 mL) was stirred at 120 °C for 6h. After this time the reaction was cooled down to RT and H2O was added. The mixture was then extracted with AcOEt 3 times. The combined organic fractions were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by FC on NH column (eluting from 100% of DCM to DCM/MeOH
95:5) affording the product of formula (4-methyl-2-phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4- triazol-3-yl)-2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone as mixture of diasteroisomers
Yield: 58 mg
LC-MS: m/z 473.59 (MH+)
The mixture of diasteroisomers was then separated into single enantiomers by chiral semipreparative HPLC Column Whelk 01 (R,R) (25 x 0.46 cm), 10 u
Mobile phase n-Hexane/Ethanol 65/35 % v/v
Preparative chiral
Flow rate (ml/min) 37 ml/min chromatography
DAD detection 220 nm protocol:
Loop 1000 μL
Total amount 58 mg
Injection 14 mg (each injection)
Diasteroisomer 1
Enantiomer 1, Yield: 11 Rt 11.66 min 100% ee mg
LC-MS: m/z 473.29 (MH+)
(2S, 3R) or (2R, 3S) or (2R, 3R) or (2S, 3S) (4-methyl-2- phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-
2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone
Figure imgf000267_0002
(2S, 3R) or (2R, 3S) or (2R, 3R) or
(2S, 3S) Example 125a
Enantiomer 2, Yield: 12 Rt 16.82 min 100% ee mg
Figure imgf000267_0001
(2S, 3R) or (2R, 3S) or (2R, 3R) or LC-MS: m/z 473.29 (MH+)
(2S, 3S)
(2S, 3R) or (2R, 3S) or (2R, 3R) or (2S, 3S) (4-methyl-2- phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-
2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone
Example 125b
NMR (400 MHz, DMSO-d6) δ 13.72 (s, 1H), 7.78 - 7.47 (m, 1H), 7.44 - 7.39 (m, 1H), 7.37 - 7.23
(m, 4H), 7.23 - 7.12 (m, 1H), 7.06 - 6.91 (m, 1H), 5.94 - 4.37 (m, 1H), 3.78 - 3.38 (m, 4H), 3.23 -
2.57 (m, 5H), 2.41 - 2.31 (m, 3H), 2.28 - 2.07 (m, 4H), 2.05 - 1.81 (m, 2H), 1.77 - 1.22 (m, 2H), 1.03
- 0.94 (m, 4H), 0.87 - 0.62 (m, 2H).
Diasteroisomer 2
Enantiomer 1, Yield: 7 Rt 17.85 min 100% ee mg
LC-MS: m/z 473.29 (MH+)
(2S, 3R) or (2R, 3S) or (2R, 3R) or (2S, 3S) (4-methyl-2- phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-
2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone
Figure imgf000268_0001
(2S, 3R) or (2R, 3S) or (2R, 3R) or
Example 125c
(2S, 3S)
Enantiomer 2, Yield: 5 Rt 25.47 min 100% ee mg LC-MS: m/z 473.29 (MH+)
(2S, 3R) or (2R, 3S) or (2R, 3R) or (2S, 3S) (4-methyl-2- phenylpiperazin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-
2-(3-propan-2-ylpyrrolidin-l-yl)phenyl]methanone
Example 125d
Figure imgf000269_0001
(2S, 3R) or (2R, 3S) or (2R, 3R) or
(2S, 3S)
1H NMR (400 MHz, DMSO-d6) δ 13.73 (bs, 1H), 7.86 - 7.45 (m, 2H), 7.42 - 7.21 (m, 5H), 7.19 - 6.82
(m, 1H), 5.98 - 4.31 (m, 1H), 3.55 - 3.38 (m, 3H), 3.24 - 2.58 (m, 5H), 2.46 - 2.26 (m, 5H), 2.22 -
2.07 (m, 3H), 2.05 - 1.74 (m, 2H), 1.67 - 1.53 (m, 1H), 1.45 - 1.32 (m, 1H), 1.07 - 0.95 (m, 3H), 0.91
- 0.65 (m, 3H).

Claims

What is claimed:
1. A compound of Formula (l-A), or a pharmaceutically acceptable salt thereof:
Figure imgf000270_0001
wherein: X1 is selected from the group of:
Figure imgf000270_0002
Figure imgf000271_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000271_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000272_0001
the wavy line
Figure imgf000272_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, S, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3;
R2 is selected from the group of carbocycle and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a carbon atom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl,
-C(=O)OH, -C(=O)-O-C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -
(CH2)n1-heterocyclyl (containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, S, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl;
R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: ee) hydrogen, halogen, cyano, or OH; ff) -CO2H or -CO2-(C1-C6 alkyl); gg) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; hh) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; ii) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and jj) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
2. A compound of Formula (l-B), or a pharmaceutically acceptable salt thereof:
Figure imgf000275_0001
wherein: X1 is selected from the group of:
Figure imgf000275_0002
Figure imgf000276_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000276_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000277_0001
the wavy line
Figure imgf000277_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, S, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: kk) hydrogen, halogen, cyano, or OH;
II) -CO2H or -CO2-(C1-C6 alkyl); mm) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; nn) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH;
00) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and pp) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
3. A compound of Formula (l-C), or a pharmaceutically acceptable salt thereof:
Figure imgf000280_0001
wherein: X1 is selected from the group of:
Figure imgf000280_0002
Figure imgf000281_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000281_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000281_0003
the wavy line
Figure imgf000282_0001
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, 5, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is 5; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd 2, 3, 5, or 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH,
CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: qq) hydrogen, halogen, cyano, or OH; rr) -CO2H or -CO2-(C1-C6 alkyl); ss) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; tt) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; uu) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and vv) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon,
N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl; R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl;
R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
4. A compound of Formula (l-D), or a pharmaceutically acceptable salt thereof:
Figure imgf000284_0001
wherein: X1 is selected from the group of:
Figure imgf000285_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000285_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000286_0001
the wavy line
Figure imgf000286_0002
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, 5, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is S; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: ww) hydrogen, halogen, cyano, or OH; xx) -CO2H or -CO2-(C1-C6 alkyl); yy) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; zz) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; aaa) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and bbb) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon, N, O, and 5; R4 is present one or two times and is independently selected from the group of H, halogen, oxo, C1-C6 alkyl, -(CH2)n2-C3-C6 cycloalkyl, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)-O-C1-C6 alkyl,
-S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), phenyl, benzyl, or a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1- C3 alkyl; and, when Y2 is carbon, R4 may also be -O-C1-C6 alkyl or two R4 may form a spirocyclic carbocycle or spirocyclic heterocycle; wherein the R4 C1-C6 alkyl, -C(=O)-O-C1-C6 alkyl, and -O-C1-C6 alkyl groups and the rings of the -(CH2)n2-C3-C6 cycloalkyl and -O-(CH2)n2-C3-C6 cycloalkyl, phenyl, and benzyl groups are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3,
OH, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and S, substituted or unsubstituted phenyl, and -O-C1-C3 alkyl;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3.
5. A compound of Formula (l-E), or a pharmaceutically acceptable salt thereof:
Figure imgf000289_0001
wherein: X1 is selected from the group of:
Figure imgf000289_0002
Figure imgf000290_0001
na is an integer selected from the group of 0, 1, and 2; nb is an integer selected from the group of 0, 1, 2, 3, and 4; with the proviso that the sum of na + nb is not less than 2 and not greater than 4; or X1 and Z1 together form a fused ring system of the formula (la):
Figure imgf000290_0002
Ra is selected from the group of hydrogen and C1-C3 alkyl;
X2 is selected from the group of:
Figure imgf000290_0003
the wavy line
Figure imgf000291_0001
in each instance represents a bond through which each X1 and X2 moiety is bound; Y1 is selected from the group of C and N; Y2 is selected from the group of C, N, 5, and O, provided R4 is not present when Y2 is O and provided R4 is either not present or present one or two times when Y2 is 5; with the proviso that no more than one of Y1 and Y2 are C; nc is an integer selected from the group of 1, 2, and 3; nd is an integer selected from the group of 1, 2, and 3; with the proviso that the sum of nc + nd is not less than 2 and not greater than 6; Z1, Z2, and Z3 are each independently selected from the group of C and N, with the proviso that Z1, Z2, and Z3, when bound to R2 or non-hydrogen R2' are C; R1 is selected from the group of C1-C6 alkyl, -(CH2)n1-C3-C6 cycloalkyl, -NRxRy,; phenyl, and benzyl, wherein the C1-C6 alkyl group and the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH,
CF3, and -O-C1-C3 alkyl, and wherein each of Rxand Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen,
OH, CF3, and -O-C1-C3 alkyl; n1 is an integer selected from the group of 0, 1, 2, and 3; R2 is selected from the group of phenyl, pyridinyl bound through a carbon atom, and a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system bound through a nitrogen heteroatom and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 0, 1, 2, 3, or 4 additional ring heteroatoms selected from the group of N, S, and O, with any of the foregoing substituted by 0, 1, 2, or 3 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, -
(CH2)n1-C3-C6 cycloalkyl, -CF3, halogen, oxo, cyano, -O-(CH2)n2-C3-C6 cycloalkyl, -C(=O)OH, -C(=O)-O- C1-C6 alkyl, -S(=O)2-C1-C6 alkyl, -C(=O)-NRxRy,; -N(Rx)(S(=O)2-C1-C6 alkyl), -(CH2)n1-heterocyclyl
(containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O), and phenyl, with the proviso that R2 is not pyridinyl unless substituted at least once by -(CH2)n1-C3-C6 cycloalkyl or phenyl; R2' is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3; R3 is present one or more times and is independently selected from the group of: ccc) hydrogen, halogen, cyano, or OH; ddd) -CO2H or -CO2-(C1-C6 alkyl); eee) C1-C6 alkyl or -O-C1-C6 alkyl substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, CF3, and OH; fff) phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3-C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, cyano, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH; ggg) a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, 5, and N, the 5-membered or 6- membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, benzyl, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH; and hhh) two R3 form a bridge between carbon atoms, wherein the atoms of the bridge comprise 1, 2, or 3 atoms independently selected from the group of carbon, N, O, and 5; R4 is present one or two times and is halogen;
R5 is selected from the group of H and C1-C6 alkyl, wherein the R5 C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -
NRxRy,; and OH, wherein each of Rxand Ry; are independently selected from the group of H and C1- C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl; R6 is selected from the group of H, C1-C6 alkyl, a heterocyclic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5, phenyl, and benzyl, wherein the R6 C1-C6 alkyl groups is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, -NRxRy,; and OH, and the rings of the R6 phenyl and benzyl groups and the heterocyclic ring are substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of C1-C6 alkyl, -O-C1-C6 alkyl, halogen, -CF3, and OH, wherein each of Rx and Ry; are independently selected from the group of H and C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from halogen, OH, CF3, and -O-C1-C3 alkyl; and n2 in each instance is an integer selected from the group of 0, 1, 2, and 3, with the proviso that the compound is not N-[4-(4,4-difluoropiperidine-l-carbonyl)-3- pyrrolidin-l-ylphenyl]cyclopropanecarboxamide; (4,4-difluoropiperidin-l-yl)-[4-(5-methyl-4H- l,2,4-triazol-3-yl)-2-[3-(trifluoromethyl)pyrazol-l-yl]phenyl]methanone; [2-(3-cyclopropylpyrazol- l-yl)-4-(5-methyl-4H-l,2,4-triazol-3-yl)phenyl]-(4,4-difluoropiperidin-l-yl)methanone; (4,4- difluoropiperidin-l-yl)-[4-(5-methyl-4H-l,2,4-triazol-3-yl)-2-(3-propan-2-ylpyrazol-l- yl)phenyl]methanone; or N-[4-(4-fluoropiperidine-l-carbonyl)-3-pyrrolidin-l- ylphenyl)cyclopropanecarboxamide.
6. The compound of any one of claims 1-5, wherein R4 is not unsubstituted benzyl.
The compound of any one of claims 1-Error! Reference source not found., wherein R4 is not unsubstituted pyridinyl.
8. The compound of any one of claims 1-5, wherein R4 is not substituted or unsubstituted phenyl.
9. The compound of any one of claims 1-Error! Reference source not found., wherein R3 is not H.
10. A compound of any one of Claims 1-9 or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000293_0001
wherein X1, X2, R2, are as defined in any one of Claims 1-9; wherein Z1, Z2, and Z3 are C; and wherein R2' is R2'a and is selected from the group of OH, halogen, C1-C6 alkyl, and -CF3.
11. A compound of any one of Claims 1-9 or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000294_0001
wherein X1, X2, R2, are as defined in any one of Claims 1-9; wherein Z1, Z2, and Z3 are C; and wherein R2' is R2'band is selected from the group of OH, halogen, C1-C6 alkyl, and -CF3.
12. A compound of any one of Claims 1-9 or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000294_0002
wherein X1, X2, R2, are as defined in any one of Claims 1-9; wherein Z1, Z2, and Z3 are C; and wherein R2' is R2'cand is selected from the group of hydrogen, OH, halogen, C1-C6 alkyl, and -CF3.
13. A compound of any one of Claims 1, 2, and 4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000295_0001
wherein, X1, Z1, Z2, Z3, Y1, Y2, R2, R2', R3, and R4 are as defined in Claim 1, 2, or 4.
14. A compound of any one of Claims 1, 2, and 4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000295_0003
wherein, X1, Z1, Z2, Z3, R2, R2', and R3 are as defined in Claim 1, 2, or 4.
15. A compound of any one of Claims 1, 2, and 4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000295_0002
wherein, X1, Z1, Z2, Z3, R2, R2', and R3 are as defined in Claim 1, 2, or 4.
16. A compound of any one of Claims 1, 2, and 4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000296_0001
wherein, X1, Z1, Z2, Z3, R2, R2', and R4 are as defined in Claim 1, 2, or 4.
17. A compound of any one of 1, 2, and 4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000296_0002
wherein, X1, Z1, Z2, Z3, R2, R2', R4, R5, and R6 are as defined in Claim 1, 2, or 4.
18. A compound of any one of Claims 1-4, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000296_0003
wherein X1, Z1, Z2, Z3, R2, R2', and R4 are as defined in one of Claims 1-4.
19. A compound of any one of Claims 1-5, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000297_0001
wherein X1, Z1, Z2, Z3, R2, R2', and R4 are as defined in one of Claims 1-5.
20. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000297_0002
wherein, Z1, Z2, Z3, Y1, Y2, Ra, R1, R2, R2', R3, and R4 are as defined in Claim 1 or 2.
21. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000297_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
22. A compound of any one of Claims 11 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000298_0001
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
23. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000298_0002
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
24. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000298_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
25. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000299_0001
wherein, Z1, Z2, Z3, Y1, Y2, Ra, R1, R2, R2', R3, and R4 are as defined in Claim 1 or 2.
26. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000299_0002
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
27. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000299_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
28. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000300_0001
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
29. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000300_0002
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
30. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000300_0003
wherein, Z1, Z2, Z3, Y1, Y2, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
31. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000301_0001
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
32. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000301_0002
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
33. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000301_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
34. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000302_0001
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
35. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000302_0002
wherein, Z1, Z2, Z3, Y1, Y2, Ra, R1, R2, R2', R3, and R4 are as defined in Claim 1 or 2.
36. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000302_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
37. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000303_0001
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
38. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000303_0002
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
39. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000303_0003
wherein, Z1, Z2, Z3, Ra, R1, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
40. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000304_0001
wherein, Z1, Z2, Z3, Y1, Y2, R1, R2, R2', R3, and R4 are as defined in Claim 1 or 2.
41. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000304_0003
wherein, Z1, Z2, Z3, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
42. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000304_0002
wherein, Z1, Z2, Z3, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
43. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000305_0001
wherein, Z1, Z2, Z3, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
44. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000305_0002
wherein, Z1, Z2, Z3, Y1, Y2, R1, R2, R2', R3, and R4 are as defined in Claim 1 or 2.
45. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000305_0003
wherein, Z1, Z2, Z3, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
46. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000306_0001
wherein, Z1, Z2, Z3, R1, R2, R2', and R4 are as defined in Claim 1 or 2.
47. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000306_0002
wherein, Z1, Z2, Z3, R1, R2, R2', and R3 are as defined in Claim 1 or 2.
48. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000306_0003
wherein, Z1, Z2, Z3, R1, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
49. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000307_0001
wherein, Z1, Z2, Z3, Y1, Y2, R2, R2', R3, R4, na, and nb are as defined in Claim 1 or 2.
50. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000307_0002
wherein, Z1, Z2, Z3, R2, R2', R3, na, and nb are as defined in Claim 1 or 2.
51. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000307_0003
wherein, Z1, Z2, Z3, R2, R2', R4, na, and nb are as defined in Claim 1 or 2.
52. A compound of any one of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000308_0002
wherein, Z1, Z2, Z3, R2, R2', R3, na, and nb are as defined in Claim 1 or 2.
53. A compound of any of Claims 1 and 2, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000308_0001
wherein, Z1, Z2, Z3, R2, R2', R5, and R6 are as defined in Claim 1 or 2.
54. A compound of any one of Claims 1, 2, 3, and 5, or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000308_0003
wherein Z1, Z2, Z3, R2, R2', and X2 are as defined in Claim 1, 2, 3, or 5.
55. A compound of any one of Claims 1, 2, 3, and 5 or a pharmaceutically acceptable salt thereof, having the formula:
Figure imgf000309_0001
wherein Z1, Z2, Z3, R2, R2', and X2 are as defined in Claim 1, 2, 3, or 5.
56. The compound of any one of Claims 1, 2, 3, and 5 , or a pharmaceutically acceptable salt thereof having the formula:
Figure imgf000309_0002
wherein Z1, Z2, Z3, R2', and X2 are as defined in one of Claims 1, 2, 3, or 5 ; and wherein R7 is selected from the group of -OH; halogen; -C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -
(CH2)n1- heterocycle (containing a monocyclic heterocyclic ring or a bicyclic or spirocyclic heterocyclic ring system and containing 3, 4, 5, 6, 7, or 8 ring carbon atoms and 1, 2, 3, or 4 ring heteroatom selected from the group of N, 5, and O); cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -C(=O)OH;
-C(=O)-O-C1-C6 alkyl; -S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
57. The compound of any of Claims any one of Claims 1, 2, 3, and 5 , or a pharmaceutically acceptable salt thereof having the formula:
Figure imgf000310_0001
wherein Z1, Z2, Z3, R2', and X2 are as defined in one of Claims 1, 2, 3, or 5; and wherein R7 is selected from the group of -OH; halogen; -C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -C(=O)OH; -C(=O)-O-C1-C6 alkyl; -S(=O)2-C1-C6 alkyl; — C(=O)-
NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
58. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1, Z2, and Z3 are each C.
59. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein one of Z1, Z2, and Z3 is N and the others C.
60. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein two of Z1, Z2, and Z3 are N and the other C.
61. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1, Z2, and Z3 are each N.
62. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1 and Z2 are each C and Z3 is N or wherein Z1 is N and Z2 and Z3 are C.
63. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1 and Z3 are each C and Z2 is N.
64. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1 and Z3 are each N and Z2 is C.
65. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z1 and Z2 are each N and Z3 is C.
66. The compound of any of Claims 1-57, or a pharmaceutically acceptable salt thereof, wherein Z2 and Z3 are each N and Z1 is C.
67. The compound of any one of Claims 1 and 6-66, or a pharmaceutically acceptable salt thereof, wherein Rg is selected from the group of:
Figure imgf000311_0001
Figure imgf000312_0001
wherein R7 is present one or two times and is independently selected from the group of hydrogen,
-OH; halogen; -C1-C6 alkyl; -O-C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -
C(=O)OH; -C(=O)-O- C1-C6 alkyl; -S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy; -N(Rx)(S(=O)2- C1-C6 alkyl); and -
CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
68. The compound of any one of Claims 1 and 6-66, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group of:
Figure imgf000313_0002
69. The compound of any one of Claims 2-66, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group of:
Figure imgf000313_0001
Figure imgf000314_0001
wherein R5 and R6 are each independently selected from hydrogen, -OH; halogen; -C1-C6 alkyl; -O- C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -C(=O)OH; -C(=O)-O-C1-C6 alkyl; -
S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
70. The compound of any one of Claims 2-66, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group of:
Figure imgf000314_0002
Figure imgf000315_0002
wherein R7 and R8 are each independently selected from hydrogen, -OH; halogen; -C1-C6 alkyl; -O-C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -C(=O)OH; -C(=O)-O-C1-C6 alkyl; -
S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy; -N(Rx)(S(=O)2-C1-C6 alkyl); and -CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
71. The compound of any one of Claims 2-66, or a pharmaceutically acceptable salt thereof, wherein R2 is selected from the group of:
Figure imgf000315_0001
wherein R5 and R6 are each independently selected from hydrogen, -OH; halogen; -C1-C6 alkyl; -O-C1-C6 alkyl; -(CH2)n1-C3-C6 cycloalkyl; -(CH2)n1-C3-C6 heterocycle including at least one heteroatom selected from O, N, and S; cyano; -O-(CH2)n2-C3-C6 cycloalkyl; -C(=O)OH; -C(=O)-O-C1-C6 alkyl; -
S(=O)2-C1-C6 alkyl; -C(=O)-NRxRy;; -N(Rx)(S(=O)2-C1-C6 alkyl); and -CF3, wherein any of the alkyl groups are further substituted with 0, 1, 2, 3, or 4 substituents selected from halogen and -OH, and wherein n1 is an integer selected from the group of 0, 1, 2, and 3.
72. The compound of any one of Claims 2-66, or a pharmaceutically acceptable salt thereof, wherein R2 is the group of:
Figure imgf000316_0001
wherein R5 and R6 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
73. The compound of any one of Claims 2-66, or a pharmaceutically acceptable salt thereof, R2 is selected from the group of:
Figure imgf000316_0002
wherein R5 and R6 are each independently selected from hydrogen, halogen, C1-C6 alkyl, -O-C1-C6 alkyl, and -CF3.
74. The compound of any of Claims 1-73, or a pharmaceutically acceptable salt thereof, wherein R5 and R6 are each independently selected from the group of hydrogen, C1-C4 alkyl, and C1-C4 alkoxy.
75. The compound of any of Claims 1-74, or a pharmaceutically acceptable salt thereof, wherein R5 and R6 are each independently selected from the group of hydrogen, C1-C3 alkyl, and C1-C3 alkoxy.
76. The compound of any of Claims 1-75, or a pharmaceutically acceptable salt thereof, wherein R3 is present 1-3 times, is present 2 times, or is present only 1 time; and
OPTIONALLY either wherein R3 is selected from the group of hydrogen, C1-C4 alkyl, C3-C6 cycloalkyl, -CF3, -C1-C4 alkyl-OH, phenyl, benzyl, pyrazolyl, and thiophenyl; wherein the phenyl, pyrazolyl, and thiophenyl rings are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, C1-C4 alkyl, and C1-C4 alkoxy; or wherein R3 is selected from the group of phenyl, benzyl, pyrazolyl, and thiophenyl, substituted by 0, 1,
2, or 3 substituents selected from the group of halogen, OH, CF3, C1-C4 alkyl, and C1-C4 alkoxy.
77. The compound of any of Claims 1-76, or a pharmaceutically acceptable salt thereof, wherein Y1 and Y2 are each N.
78. The compound of any of Claims 1-76, or a pharmaceutically acceptable salt thereof, wherein Y1 is N and Y2 is C.
79. The compound of any of Claims 1-76, or a pharmaceutically acceptable salt thereof, wherein Y1 is C and Y2 is N.
80. The compound of any of Claims 1-79, or a pharmaceutically acceptable salt thereof, wherein R1 is C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl.
81. The compound of any of Claims 1-79, or a pharmaceutically acceptable salt thereof, wherein R1 is C1-C4 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -O-C1-C3 alkyl.
82. The compound of any of Claims 1-79, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from the group of -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl, wherein the rings of the -(CH2)n1-C3-C6 cycloalkyl, phenyl, and benzyl groups are substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, OH, CF3, and -
O-C1-C3 alkyl.
83. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein Ra is H.
84. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein Ra is C1-C3 alkyl.
85. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3 is H.
86. The compound of any of Claims the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3 is C1-C3 alkyl.
87. The compound of any of Claims the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group of CO2H and -CO2-(C1-C6 alkyl).
88. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group of phenyl, benzyl, C3-C6 cycloalkyl, and -
CH2-C3-C6 cycloalkyl, the rings of each of the phenyl, benzyl, C3-C6 cycloalkyl, and -CH2-C3- C6 cycloalkyl groups being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen, -CF3, and OH.
89. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from the group of a 5-membered or 6-membered heterocyclic ring containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, the 5-membered or 6-membered heterocyclic ring being substituted by 0, 1, 2, or 3 substituents selected from OH, halogen, and C1-C6 alkyl, wherein the C1-C6 alkyl group is further substituted by 0, 1, 2, 3, 4 or 5 substituents selected from the group of halogen and OH.
90. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the sum of na + nb is 3.
91. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the sum of na + nb is 4.
92. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is C1-C6 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl.
93. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is C1-C4 alkyl substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl.
94. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is C1-C4 alkyl substituted by 0, 1, 2, or 3 halogen and OH.
95. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is -(CH2)n2-C3-C6 cycloalkyl, wherein the cycloalkyl ring of the -(CH2)n2- C3-C6 cycloalkyl group is substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl: and n2 is an integer selected from the group of 0, 1,
2, and 3.
96. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from the group of phenyl and benzyl, with the rings of the phenyl and benzyl groups being substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl.
97. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R4 is a heterocyclic or heteroaromatic ring having 3, 4, 5, 6, 7, 8, 9, or 10 ring atoms of which 1, 2, 3, or 4 ring atoms are selected from the group of N, O, and 5; wherein the rings of the phenyl and benzyl groups are each independently substituted by
0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl.
98. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein Y2 is C and EITHER R4 is selected from the group of H, -O-C1-C6 alkyl, and -
O-(CH2)n2-C3-C6 cycloalkyl; wherein the O-C1-C6 alkyl group and the cycloalkyl ring of the -
O-(CH2)n2-C3-C6 cycloalkyl are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen, CF3, OH, and -O-C1-C3 alkyl OR two R4 form a C3-C6 cycloalkyl carbocycle or heterocycle containing 2-5 ring carbon atoms and 1 or 2 ring heteroatoms selected from the group of N and O.
99. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein the -C1-C6 alkyl group and the cycloalkyl ring of the -O-(CH2)n2-C3-C6 cycloalkyl are each independently substituted by 0, 1, 2, or 3 substituents selected from the group of halogen and OH.
100. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound selected from any of the preceding claims, a compound selected from
Examples 1-125, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient.
101. The use of a compound selected from any of the preceding claims, a compound selected from Examples 1-125, or a pharmaceutically acceptable salt of any of the foregoing, in the preparation of a medicament.
102. A compound selected from any of the preceding claims, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing, for use as a medicament.
103. A method of inhibiting the activity of a GPR39 protein in a subject, the method comprising administering to the subject in need thereof a pharmaceutically effective amount of a compound selected from any of the preceding claims, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
104. A method of treating hypertension in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
105. The method of Claim 104, wherein the hypertension is primary hypertension.
106. The method of Claim 104, wherein the hypertension is secondary hypertension.
107. The method of Claim 104, wherein the hypertension is treatment-resistant or refractory hypertension.
108. The method of Claim 104, wherein the hypertension is pulmonary hypertension.
109. A method of treating heart failure in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
110. A method of breast cancer in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of
Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
111. A method of gastric adenocarcinomas in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
112. A method of promoting or enhancing colon epithelial function and tight junction barrier integrity in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims
1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
113. A method of treating ulcerative colon disease in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
114. The method of Claim 113, wherein the ulcerative colon disease is ulcerative colitis.
115. The method of Claim 113, wherein the ulcerative colon disease is Crohn's Disease.
116. A method of treating Inflammatory Bowel Disease in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from
Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
117. A method of treating diarrhea in a human, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
118. A method of enhancing the delivery of an anesthetic to a human experiencing microvascular complications, the method comprising administering to the human in need thereof a pharmaceutically effective amount of a compound selected from any of Claims
1-102, a compound selected from Examples 1-125, a compound of Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically effective amount of the anesthetic.
119. The method of Claim 118, wherein the human is experiencing microvascular complications associated with or caused by prediabetes.
120. The method of Claim 118, wherein the human is experiencing microvascular complications associated with or caused by diabetes.
121. A method of treating stroke in a human in need thereof, the method comprising administering to the human a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, a compound of
Formula (I'), or a pharmaceutically acceptable salt of any of the foregoing.
122. A kit comprising: a) one or more compositions, each composition comprising a pharmaceutically effective amount of a compound selected from any of Claims 1-102, a compound selected from Examples 1-125, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient; and b) instructions for administering the one or more compositions to a human in need thereof.
123. A compound of any one of Examples 1-125, or a pharmaceutically acceptable salt thereof.
PCT/US2022/047658 2021-10-26 2022-10-25 Antagonists of gpr39 protein WO2023076219A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163272076P 2021-10-26 2021-10-26
US63/272,076 2021-10-26

Publications (2)

Publication Number Publication Date
WO2023076219A2 true WO2023076219A2 (en) 2023-05-04
WO2023076219A3 WO2023076219A3 (en) 2023-06-08

Family

ID=84362369

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/047658 WO2023076219A2 (en) 2021-10-26 2022-10-25 Antagonists of gpr39 protein

Country Status (1)

Country Link
WO (1) WO2023076219A2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040071708A1 (en) 2002-09-26 2004-04-15 Immusol, Inc. GPR 39 modulators that control cancerous cell growth
WO2007141322A1 (en) 2006-06-08 2007-12-13 Janssen Pharmaceutica N.V. G protein coupled receptor 39 (gpr39)
US20090298756A1 (en) 2005-04-22 2009-12-03 Shanghai Institutes For Biological Sciences, Chinese Academy Of Sciences Functions and uses of gpr39 gene in mammalian central nervous system

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6924302B2 (en) * 2000-12-22 2005-08-02 Ortho Mcneil Pharmaceutical, Inc. Substituted triazole diamine derivatives as kinase inhibitors
US20140010783A1 (en) * 2012-07-06 2014-01-09 Hoffmann-La Roche Inc. Antiviral compounds
US20150376198A1 (en) * 2013-02-18 2015-12-31 The Scripps Research Institute Modulators of vasopressin receptors with therapeutic potential
AR114136A1 (en) * 2017-10-10 2020-07-29 Hoffmann La Roche HETEROCYCLIC COMPOUNDS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040071708A1 (en) 2002-09-26 2004-04-15 Immusol, Inc. GPR 39 modulators that control cancerous cell growth
US20090298756A1 (en) 2005-04-22 2009-12-03 Shanghai Institutes For Biological Sciences, Chinese Academy Of Sciences Functions and uses of gpr39 gene in mammalian central nervous system
WO2007141322A1 (en) 2006-06-08 2007-12-13 Janssen Pharmaceutica N.V. G protein coupled receptor 39 (gpr39)

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"GIfunctions of GPR39: novel biology", CURRENT OPINION IN PHARMACOLOGY, vol. 12, 2012, pages 647 - 652
ALEN ET AL., ONCOTARGET, vol. 7, 2016, pages 5957 - 5971
COHEN ET AL.: "The zinc sensing receptor, ZnR/GPR39, controls proliferation and differentiation of colonocytes and thereby tight junction formation in the colon", CELL DEATH AND DISEASE, vol. 5, 2014, pages e1307
MOECHARS ET AL.: "Altered Gastrointestinal and Metabolic Function in the GPR39-Obestatin Receptor-Knockout Mouse", GASTROENTEROLOGY, vol. 131, October 2006 (2006-10-01), pages 1131 - 1141, XP005821163, DOI: 10.1053/j.gastro.2006.07.009
VENTURA-BIXENSHPANER ET AL.: "Enhanced ZnR/GPR39 Activity in Breast Cancer, an Alternative Trigger of Signaling Leading to Cell Growth", SCIENTIFIC REPORTS, vol. 8, 2018, pages 8119

Also Published As

Publication number Publication date
WO2023076219A3 (en) 2023-06-08

Similar Documents

Publication Publication Date Title
KR101799007B1 (en) 1,3,4-Oxadiazole Sulfonamide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
AU2018200536B2 (en) Amino pyrazine derivatives as phosphatidylinositol 3-kinase inhibitors
TWI789381B (en) Heterocyclic compound
RU2610840C2 (en) Thiazolpyrimidines
AU2018226485A1 (en) Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of Hepatitis B
JP6231085B2 (en) Aminoquinazoline derivatives and pyridopyrimidine derivatives
US8940716B2 (en) Bicyclic heteroaryl compounds as GPR119 modulators
AU2017380282B2 (en) Pyran derivatives as cyp11a1 (cytochrome p450 monooxygenase 11a1) inhibitors
AU2016231832A1 (en) Novel 3-indol substituted derivatives, pharmaceutical compositions and methods for use
CN105682661A (en) Certain chemical entities, compositions, and methods
WO2016126722A1 (en) 3-alkyl bicyclic [4,5,0] hydroxamic acids as hdac inhibitors
CA2962578A1 (en) Aminotriazine derivatives useful as tank-binding kinase inhibitor compounds
EP2809652B1 (en) Isoquinoline and naphthyridine derivatives
JP2023508930A (en) TRPML modulator
KR102606799B1 (en) Poly-ADP ribose polymerase (PARP) inhibitor
CA3199496A1 (en) Heterocycle derivatives for treating trpm3 mediated disorders
AU2021288584A1 (en) Collagen 1 translation inhibitors and methods of use thereof
WO2023076219A2 (en) Antagonists of gpr39 protein
CN117957219A (en) Pyridine derivative and application thereof
CN116870016A (en) Heteroaromatic compound and medical application thereof
JP2024511681A (en) N2-phenylpyrimidine-2,4-diamine compounds and their preparation and use methods
US20230174484A1 (en) Antagonists of gpr39 protein
KR20230153959A (en) Pim kinase inhibitors and methods of their use
JPWO2021222858A5 (en)