WO2017061957A1 - Compounds for treatment of cancer and epigenetics - Google Patents

Compounds for treatment of cancer and epigenetics Download PDF

Info

Publication number
WO2017061957A1
WO2017061957A1 PCT/SG2016/050499 SG2016050499W WO2017061957A1 WO 2017061957 A1 WO2017061957 A1 WO 2017061957A1 SG 2016050499 W SG2016050499 W SG 2016050499W WO 2017061957 A1 WO2017061957 A1 WO 2017061957A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
methyl
optionally substituted
pyridinyl
fluoro
Prior art date
Application number
PCT/SG2016/050499
Other languages
French (fr)
Inventor
Klement Jihao FOO
Anders Poulsen
Thomas Hugo Keller
Si SI LIEW
Cheng San Brian Chia
Jin Yan Melgious ANG
Chuhui Huang
Original Assignee
Agency For Science, Technology And Research
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 Agency For Science, Technology And Research filed Critical Agency For Science, Technology And Research
Priority to CA3001452A priority Critical patent/CA3001452A1/en
Priority to CN201680071788.2A priority patent/CN108419435A/en
Priority to SG11201802952TA priority patent/SG11201802952TA/en
Priority to JP2018518408A priority patent/JP2018534276A/en
Priority to AU2016335043A priority patent/AU2016335043A1/en
Priority to KR1020187012151A priority patent/KR20180061316A/en
Priority to BR112018007128A priority patent/BR112018007128A2/en
Priority to EP16854005.2A priority patent/EP3371151A4/en
Priority to US15/767,129 priority patent/US20190071416A1/en
Publication of WO2017061957A1 publication Critical patent/WO2017061957A1/en

Links

Classifications

    • 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
    • C07D401/06Heterocyclic 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 only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D219/00Heterocyclic compounds containing acridine or hydrogenated acridine ring systems
    • C07D219/04Heterocyclic compounds containing acridine or hydrogenated acridine ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/16Ring systems of three rings containing carbocyclic rings other than six-membered
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D413/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D413/06Heterocyclic 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 only aliphatic carbon atoms
    • 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
    • C07D413/12Heterocyclic 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 chain containing hetero atoms as chain links
    • 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
    • C07D417/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • 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
    • C07D471/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention generally relates to quinolines and 5,6,7,8-tetrahydroacridine derivatives, methods for their preparation, pharmaceutical compositions containing these compounds and uses of these compounds in the treatment of disorders/conditions/diseases involving, relating to or associated with enzymes having methyltransferase activities.
  • chromatin forms the structural basis of nuclear processes associated with genetic activity.
  • Nucleosome is the smallest structural unit of chromatin, in which 146 base pairs of DNA are wrapped around an octamer of core histones.
  • the histones are subjected to several post-translational covalent modifications and this plays a critical role in controlling gene transcription within cells.
  • methylation of lysine residue seems to play a particularly important role in control of gene transcription programs (Arrowsmith, C. H., et al. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 2012, 11, 384-400).
  • PKMT protein lysine methyltransferases
  • PKMTs contain an evolutionarily conserved SET (Su(var), E(z) and Trithorax) domain that is involved in catalyzing the transfer of the methyl group from the cofactor 5- adenosyl-L-methionine (SAM) to lysine residues of histone and non-histone substrates, leading to lysine mono-, di-, and/or trimethylation.
  • SAM 5- adenosyl-L-methionine
  • Histone lysine methylation has been increasingly recognized as a major epigenetic gene regulation mechanism in eukaryotic cells (Copeland, R. A., Molecular pathways: protein methyltransferases in cancer. Clin. Cancer Res. 2013, 19, 6344-6350).
  • SMYD3 is a histone methyltransferase and is overexpressed in several cancers including breast, gastric, pancreatic, colorectal, lung cancer and hepatocellular carcinoma. It tri-methylates histone H3 at lysine 4 (H3K4me3), a mark associated with gene activation.
  • H3K4me3 histone H3 at lysine 4
  • SMYD3 is part of the SMYD family (SET/MYND) of proteins which contains five members carrying a SET domain and a MYND type of zinc finger.
  • the up-regulation of the SMYD3 promotes proliferation of HCC (hepatocellular carcinoma) via increasing H3 lysine 4 methylation, and a subsequent activation of downstream genes, including Nkx2.8 gene which is frequently up-regulated in human HCC (Hamamoto, R., et al. SMYD3 encodes a histone methyltransf erase involved in the proliferation of cancer cells. Nat. Cell Biol. 2004, 6, 731-740).
  • SMYD3 also catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of SMYD3 protein. It has been shown that the catalytic activity of SMYD3 is required for the anchorage-independent growth of cancer cells. Thus, SMYD3, via H4K5 methylation, provides another link between chromatin dynamics and neoplastic disease (Van Eller, et al. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics 2012, 7, 340-343).
  • SMYD3 modulates myostatin and c-Met transcription in primary skeletal muscle cells and C2C12 myogenic cells. It does this by targeting the myostatin and c-Met genes and participates in the recruitment of the bromodomain protein BRD4 to their regulatory regions through protein-protein interaction. By recruiting BRD4, SMYD3 favors chromatin engagement of the pause-release factor p- TEFb (positive transcription elongation factor) and elongation of Ser2-phosphorylated RNA polymerase II (PolIISer2P). SMYD3 is also known to methylate other substrates such as RBI protein (CA2613322 Al) and VEGFR1 (US8354223 B2).
  • SMYD3 acts in the cytoplasm of cancer cells, methylating a lysine residue (K260) on MAP3K2, a kinase enzyme that is associated with the activation of MEK-ERK mitogen-activated protein-kinase pathway (Mazur, P., et.al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature 2014, 510, 283- 287).
  • tetrahydroacridine derivatives found to act against against a different target ubiquitin specific protease 7.
  • the compounds disclosed in WO2011/086178 were found to act against a different target ubiquitin specific protease 7, but were not found to be active against SMYD3.
  • Z 1 and Z 2 are independently selected from O, S or NH;
  • X is a halogen
  • R 1 and R 2 are independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl , optionally substituted aryl and optionally substituted heteroaryl;
  • R 1 and R 2 may optionally be taken together to form an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S;
  • R 1 and R 2 may optionally form an optionally substituted aryl or optionally substituted heteroaryl together with the ring atoms that they are bonded to;
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently absent, or selected from the group consisting of a bond, hydrogen, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
  • R 3 , R 4 , R 5 , R 6 , R 7 or R 8 may be taken together to form an optionally substituted cycloalkyl or an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S ;
  • Y is selected from R 9 , OR 9 or NHR 9 , wherein R 9 is an optionally substituted C 3 to do alkyl, optionally substituted C 3 to do alkenyl, optionally substituted C 3 to do alkynyl, optionally substituted d to d cycloalkyl, optionally substituted d to do haloalkyl, a substituted 5-membered heteroaryl comprising two to three heteroatoms selected from N, O or S or a d to d alkyl substituted with an optionally substituted 5-membered heterocycloalkyl comprising one to two heteroatoms selected from N, O or S;
  • R 1 and R l la are independently selected from the group consisting of H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
  • R nb may be absent, H or optionally substituted alkyl
  • a 2 is selected from CH, N, O or S;
  • p is an integer selected from 0, 1 or 2.
  • a 3 and A 4 are independently selected from CH or N;
  • a 5 and A 6 are independently selected from CH, N, O or S;
  • R 12a , R 13a , R 14 and R 15 are independently selected from the group consisting of hydrogen, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
  • R 12b and R 13b are independently absent, H or an optionally substituted alkyl
  • q and r are independently integers selected from 0, 1 or 2.
  • the compound as defined above is an inhibitor of protein lysine methyltransferases (PKMT) such as SMYD3.
  • PKMT protein lysine methyltransferases
  • SMYD3 is an attractive target for drug discovery due to its role in epigenetic regulation and crucial cell signalling pathways.
  • a small molecule inhibitor of SMYD3 as defined above may be useful for the treatment of cancers with elevated SMYD3 expression such as hepatocellular carcinoma (HCC).
  • HCC hepatocellular carcinoma
  • the compounds as defined above have a unique potency profile against the target protein.
  • the compounds may be modified to have different potencies against different targets for a variety of indications or applications.
  • the compound is a small molecule inhibitor. Small molecule inhibitors, unlike macromolecules such as polymers, proteins and DNA, may be less toxic and have fewer occurrences of adverse drug effects while maintaining a high level of activity.
  • the tested compounds as defined above have a siginificantly higher potency against the target protein compared to conventionally known compounds.
  • the compound as defined above has a significantly higher potency in inhibiting SMYD3.
  • a pharmaceutical composition comprising a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, and a pharmaceutically acceptable excipient.
  • a method of inhibiting SMYD3 in a cell comprising administering to a cell the compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above.
  • a method of treating a SMYD3-related disorder comprising administering to a subject in need of treatment a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above.
  • a compound as defined above or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above, in the manufacture of a medicament for treatment of a SMYD3-related disorder.
  • a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above, for use in the treatment of a SMYD3- related disorder for use in the treatment of a SMYD3- related disorder.
  • the compounds as defined above have demonstrated inhibitory activities against the methyl transferase activity of SMYD3 enzyme and anti-proliferative activities against a variety of human tumor cell lines.
  • the compound as defined above may demonstrate good drug-like properties, that is, in vitro metabolic stability, solubility and desirable lipophilicity.
  • the compounds inhibit methyltransferase activity of SMYD3 in an MTase assay using MAP3K2 as a peptide substrate.
  • the compounds show antiproliferative activity.
  • the compounds inhibit SMYD3 mediated methylation of MAP3K2 and inhibit anchorage independent growth in human cancer cells.
  • R 16 is selected from the group consisting of H, methyl, COOMe and COOEt;
  • step (b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen
  • step (c) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form the compound of formula (III);
  • step (b) and (c) may be performed simultaneously, sequentially or in any order.
  • step a) selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form an amide;
  • step (c) selectively functionalizing at least one halogen of the halogenated cyclized product of step (a) with a group having the following formula (VII) under reaction conditions to form the compound of formula (III);
  • step (b) and (c) may be performed simultaneously, sequentially or in any order.
  • step (b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen
  • step a) optionally selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid
  • step (d) selectively functionalizing the carboxylic acid of the cyclized product of step (a) or (c) with a group having the following formula (VI) under reaction conditions to form the compound of formula (IV);
  • step (b), (c) and (d) may be performed simultaneously, sequentially or in any order.
  • alkyl As an example, some publications would use the term “alkylene” for a bridging group and hence in these other publications there is a distinction between the terms “alkyl” (terminal group) and “alkylene” (bridging group). In the present application no such distinction is made and most groups may be either a bridging group or a terminal group.
  • examples of acyl include acetyl, benzoyl and amino acid derived aminoacyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Aliphatic means non-aromatic, open chain, straight or branched organic compounds.
  • Alkenyl as a group or part of a group denotes an aliphatic hydrocarbon group containing at least one carbon -carbon double bond and which may be straight or branched preferably having 2-12 carbon atoms, more preferably 2-10 carbon atoms, most preferably 2-6 carbon atoms, in the normal chain.
  • the group may contain a plurality of double bonds in the normal chain and the orientation about each is independently E or Z.
  • Exemplary alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl and nonenyl.
  • the group may be a terminal group or a bridging group.
  • alkenyloxy refers to an alkenyl-O- group in which alkenyl is as defined herein.
  • Preferred alkenyloxy groups are C C 6 alkenyloxy groups.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Alkyl or “alkylene” as a group or part of a group refers to a straight or branched aliphatic hydrocarbon group, preferably a Q-Q 2 alkyl, more preferably a Q-Qo alkyl, most preferably C C 6 unless otherwise noted.
  • suitable straight and branched Q-Ce alkyl substituents include methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, t-butyl, hexyl, and the like.
  • the group may be a terminal group or a bridging group.
  • Alkylamino includes both mono-alkylamino and dialkylamino, unless specified.
  • Mono- alkylamino means an Alkyl-NH- group, in which alkyl is as defined herein.
  • Dialkylamino means a (alkyl) 2 N- group, in which each alkyl may be the same or different and are each as defined herein for alkyl.
  • the alkyl group is preferably a C C 6 alkyl group.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
  • Alkyloxy refers to an alkyl-O- group in which alkyl is as defined herein.
  • the alkyloxy is a C C 6 alkyloxy. Examples include, but are not limited to, methoxy and ethoxy.
  • the group may be a terminal group or a bridging group.
  • alkyloxy may be used interchangeably with the term "alkoxy”.
  • Alkyloxy alkyl refers to an alkyloxy-alkyl- group in which the alkyloxy and alkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • Alkyloxyaryl refers to an alkyloxy-aryl- group in which the alkyloxy and aryl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the aryl group.
  • the alkyl group is preferably a Q-Ce alkyl group. Examples include, but are not limited to, methoxycarbonyl and ethoxycarbonyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
  • Alkyloxycycloalkyl refers to an alkyloxy-cycloalkyl- group in which the alkyloxy and cycloalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the cycloalkyl group.
  • Alkyloxyheteroaryl refers to an alkyloxy-heteroaryl- group in which the alkyloxy and heteroaryl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroaryl group.
  • Alkyloxyheterocycloalkyl refers to an alkyloxy-heterocycloalkyl- group in which the alkyloxy and heterocycloalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heterocycloalkyl group.
  • the alkyl group is preferably a C C 6 alkyl group.
  • Exemplary alkylsulfinyl groups include, but not limited to, methylsulfinyl and ethylsulfinyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
  • the alkyl group is preferably a Q-Ce alkyl group. Examples include, but not limited to methylsulfonyl and ethylsulfonyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
  • Alkynyl as a group or part of a group means an aliphatic hydrocarbon group containing a carbon-carbon triple bond and which may be straight or branched preferably having from 2-12 carbon atoms, more preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms in the normal chain.
  • Exemplary structures include, but are not limited to, ethynyl and propynyl.
  • the group may be a terminal group or a bridging group.
  • Alkynyloxy refers to an alkynyl-O- group in which alkynyl is as defined herein. Preferred alkynyloxy groups are C C 6 alkynyloxy groups.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Amino acid as a group or part of a group means having at least one primary, secondary, tertiary or quaternary amino group, and at least one acid group, wherein the acid group may be a carboxylic, sulfonic, or phosphonic acid, or mixtures thereof.
  • the amino groups may be "alpha", “beta”, “gamma” ... to “omega” with respect to the acid group(s).
  • the amino acid may be natural or synthetic, and may include their derivatives.
  • the backbone of the "amino acid” may be substituted with one or more groups selected from halogen, hydroxy, guanido, heterocyclic groups.
  • amino acids also includes within its scope glycine, alanine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tryptophan, serine, threonine, cysteine, tyrosine, asparagine, glutamine, asparte, glutamine, lysine, arginine and histidine, taurine, betaine, N-methylalanine etc.
  • L and (D) forms of amino acids are included in the scope of this disclosure.
  • the amino acids suitable for use in the present disclosure may be derivatized to include amino acids that are hydroxylated, phosphorylated, sulfonated, acylated, and glycosylated, to name a few.
  • amino acid residue refers to amino acid structures that lack a hydrogen atom of the amino group (-NH-CHR-COOH), or the hydroxy moiety of the carboxy group (NH2-CHR-CO-), or both (- NH-CHR-CO-).
  • Amino refers to groups of the form -NR a R b wherein R a and R b are individually selected from the group including but not limited to hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, and optionally substituted aryl groups.
  • aminocarbonyl group and “carbonylamino” group can be used interchangeably and are used to describe a -CO-NR 2 group.
  • Aminoalkyl means an NH 2 -alkyl- group in which the alkyl group is as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
  • Aryl as a group or part of a group denotes (i) an optionally substituted monocyclic, or fused polycyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) preferably having from 6 to 12 atoms per ring.
  • aryl groups include phenyl, naphthyl, and the like; (ii) an optionally substituted partially saturated bicyclic aromatic carbocyclic moiety in which a phenyl and a C 5 _ 7 cycloalkyl or C 5 _ 7 cycloalkenyl group are fused together to form a cyclic structure, such as tetrahydronaphthyl, indenyl or indanyl.
  • the group may be a terminal group or a bridging group.
  • an aryl group is a C 6 -C 18 aryl group.
  • Arylalkenyl means an aryl-alkenyl- group in which the aryl and alkenyl are as defined herein. Exemplary arylalkenyl groups include phenylallyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
  • Arylalkyl means an aryl-alkyl- group in which the aryl and alkyl moieties are as defined herein. Preferred arylalkyl groups contain a C 1 5 alkyl moiety. Exemplary arylalkyl groups include benzyl, phenethyl, 1-naphthalenemethyl and 2-naphthalenemethyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • Arylalkyloxy refers to an aryl-alkyl-O- group in which the alkyl and aryl are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Arylamino includes both mono-arylamino and di-arylamino unless specified.
  • Mono-arylamino means a group of formula arylNH-, in which aryl is as defined herein.
  • Di-arylamino means a group of formula (aryl) 2 N- where each aryl may be the same or different and are each as defined herein for aryl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Arylheteroalkyl means an aryl-heteroalkyl- group in which the aryl and heteroalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
  • Aryloxy refers to an aryl-O- group in which the aryl is as defined herein.
  • the aryloxy is a Ce-Qg aryloxy, more preferably a C -C w aryloxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
  • a “bond” is a linkage between atoms in a compound or molecule.
  • the bond may be a single bond, a double bond, or a triple bond, as valency permits.
  • Cycloaliphatic means non-aromatic, cyclic organic compounds.
  • Cycloalkenyl means a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond and preferably having from 5-10 carbon atoms per ring.
  • Exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl or cycloheptenyl.
  • the cycloalkenyl group may be substituted by one or more substituent groups.
  • a cycloalkenyl group typically is a C 3 -Q 2 alkenyl group. The group may be a terminal group or a bridging group.
  • Cycloalkyl refers to a saturated monocyclic or fused or bridged or spiro polycyclic, carbocycle preferably containing from 3 to 9 carbon atoms per ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like, unless otherwise specified. It includes monocyclic systems such as cyclopropyl and cyclohexyl, bicyclic systems such as decalin, and polycyclic systems such as adamantane.
  • a cycloalkyl group typically is a C3-Q2 alkyl group. The group may be a terminal group or a bridging group.
  • Cycloalkylalkyl means a cycloalkyl-alkyl- group in which the cycloalkyl and alkyl moieties are as defined herein.
  • Exemplary monocycloalkylaikyl groups include cyclopropylmethyl, cyclopentylmethyl, cyclohexylmethyl and cycloheptylmethyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • Cycloalkylalkenyl means a cycloalkyl-alkenyl- group in which the cycloalkyl and alkenyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
  • Cycloalkylheteroalkyl means a cycloalkyl-heteroalkyl- group in which the cycloalkyl and heteroalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
  • Cycloalkyloxy refers to a cycloalkyl-O- group in which cycloalkyl is as defined herein.
  • the cycloalkyloxy is a Ci-Cecycloalkyloxy. Examples include, but are not limited to, cyclopropanoxy and cyclobutanoxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Cycloalkenyloxy refers to a cycloalkenyl-O- group in which the cycloalkenyl is as defined herein.
  • the cycloalkenyloxy is a Q-Cecycloaikenyloxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Cycloamino refers to a saturated monocyclic, bicyclic, or polycyclic ring containing at least one nitrogen atom in at least one ring. Each ring is preferably containing from 3 to 10 carbon atoms per ring, more preferably 4 to 7 carbon atoms per ring.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Haloalkyl refers to an alkyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom selected from the group consisting of fluorine, chlorine, bromine and iodine.
  • a haloalkyl group typically has the formula C n H (2n+ i m) X m wherein each X is independently selected from the group consisting of F, CI, Br and I .
  • n is typically from 1 to 10, more preferably from 1 to 6, most preferably 1 to 3.
  • m is typically 1 to 6, more preferably 1 to 3.
  • Examples of haloalkyl include fluoromethyl, difluoromethyl and trifluoromethyl.
  • Haloalkenyl refers to an alkenyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom independently selected from the group consisting of F, CI, Br and I.
  • Haloalkynyl refers to an alkynyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom independently selected from the group consisting of F, CI, Br and I.
  • Heteroalkyl refers to a straight- or branched-chain alkyl group preferably having from 2 to 12 carbon atoms, more preferably 2 to 6 carbon atoms in the chain, one or more of which has been replaced by a heteroatom selected from S, O, P and N.
  • exemplary heteroalkyls include alkyl ethers, secondary and tertiary alkyl amines, amides, alkyl sulfides, and the like.
  • heteroalkyl also include hydroxyQ-Ceaikyl, Q-CeaikyloxyQ-Ceaikyl, arninoQ-Ceaikyl, Q-CeaikylaminoQ-Ceaikyl, and di(C 1 -C 6 alkyl)aminoC 1 -C 6 alkyl.
  • the group may be a terminal group or a bridging group.
  • Heteroalkyloxy refers to a heteroalkyl-O- group in which heteroalkyl is as defined herein.
  • the heteroalkyloxy is a C C 6 heteroalkyloxy.
  • the group may be a terminal group or a bridging group.
  • Heteroaryl either alone or part of a group refers to groups containing an aromatic ring (preferably a 5 or 6 membered aromatic ring) having one or more heteroatoms as ring atoms in the aromatic ring with the remainder of the ring atoms being carbon atoms. Suitable heteroatoms include nitrogen, oxygen and sulphur.
  • heteroaryl examples include thiophene, benzothiophene, benzofuran, benzimidazole, benzoxazole, benzothiazole, benzisothiazole, naphtho[2,3-b]thiophene, furan, isoindolizine, xantholene, phenoxatine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, tetrazole, indole, isoindole, lH-indazole, purine, quinoline, isoquinoline, phthalazine, naphthyridine, quinoxaline, cinnoline, carbazole, phenanthridine, acridine, phenazine, thiazole, isothiazole, phenothiazine, oxazole, isooxazole, furazane, phen
  • a heteroaryl group is typically a Q-Qg heteroaryl group.
  • a heteroaryl group may comprise 3 to 8 ring atoms.
  • a heteroaryl group may comprise 1 to 3 heteroatoms independently selected from the group consisting of N, O and S.
  • the group may be a terminal group or a bridging group.
  • Heteroarylalkyl means a heteroaryl-alkyl group in which the heteroaryl and alkyl moieties are as defined herein. Preferred heteroarylalkyl groups contain a lower alkyl moiety. Exemplary heteroarylalkyl groups include pyridinylmethyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • Heteroarylalkenyl means a heteroaryl-alkenyl- group in which the heteroaryl and alkenyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
  • Heteroarylheteroalkyl means a heteroaryl-heteroalkyl- group in which the heteroaryl and heteroalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
  • Heteroarylamino refers to groups containing an aromatic ring (preferably 5 or 6 membered aromatic ring) having at least one nitrogen and at least another heteroatom as ring atoms in the aromatic ring, preferably from 1 to 3 heteroatoms in at least one ring. Suitable heteroatoms include nitrogen, oxygen and sulphur.
  • Arylamino and aryl is as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Heteroaryloxy refers to a heteroaryl-O- group in which the heteroaryl is as defined herein. Preferably the heteroaryloxy is a Ci-Ci 8 heteroaryloxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Heterocyclic refers to saturated, partially unsaturated or fully unsaturated monocyclic, bicyclic or polycyclic ring system containing at least one heteroatom selected from the group consisting of nitrogen, sulfur and oxygen as a ring atom.
  • heterocyclic moieties include heterocycloalkyl, heterocycloalkenyl and heteroaryl.
  • Heterocycloalkenyl refers to a heterocycloalkyl as defined herein but containing at least one double bond.
  • a heterocycloalkenyl group typically is a C 2 -Cn heterocycloalkenyl group.
  • the group may be a terminal group or a bridging group.
  • Heterocycloalkyl refers to a saturated monocyclic, fused or bridged or spiro polycyclic ring containing at least one heteroatom selected from nitrogen, sulfur, oxygen, preferably from 1 to 3 heteroatoms in at least one ring. Each ring is preferably from 3 to 10 membered, more preferably 4 to 7 membered.
  • heterocycloalkyl substituents include pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiofuranyl, piperidinyl, piperazinyl, tetrahydropyranyl, morpholino, 1 ,3- diazapane, 1 ,4-diazapane, 1 ,4-oxazepane, and 1,4-oxathiapane.
  • a heterocycloalkyl group typically is a C 2 -C 12 heterocycloalkyl group.
  • a heterocycloalkyl group may comprise 3 to 9 ring atoms.
  • a heterocycloalkyl group may comprise 1 to 3 heteroatoms independently selected from the group consisting of N, O and S. The group may be a terminal group or a bridging group.
  • Heterocycloalkylalkyl refers to a heterocycloalkyl-alkyl- group in which the heterocycloalkyl and alkyl moieties are as defined herein.
  • exemplary heterocycloalkylalkyl groups include (2-tetrahydrofuranyl)methyl, (2-tetrahydrothiofuranyl)methyl.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
  • Heterocycloalkylalkenyl refers to a heterocycloalkyl-alkenyl- group in which the heterocycloalkyl and alkenyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
  • Heterocycloalkylheteroalkyl means a heterocycloalkyl-heteroalkyl- group in which the heterocycloalkyl and heteroalkyl moieties are as defined herein.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
  • Heterocycloalkyloxy refers to a heterocycloalkyl- O- group in which the heterocycloalkyl is as defined herein.
  • the heterocycloalkyloxy is a Ci-Ceheterocycloalkyloxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Heterocycloalkenyloxy refers to a heterocycloalkenyl-O- group in which heterocycloalkenyl is as defined herein.
  • the heterocycloalkenyloxy is a C C 6 heterocycloalkenyloxy.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
  • Heterocycloamino refers to a saturated monocyclic, bicyclic, or polycyclic ring containing at least one nitrogen atom and at least another heteroatom selected from nitrogen, sulfur, oxygen, preferably from 1 to 3 heteroatoms in at least one ring.
  • Each ring is preferably from 3 to 10 membered, more preferably 4 to 7 membered.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Hydroalkyl refers to an alkyl group as defined herein in which one or more of the hydrogen atoms have been replaced with an OH group.
  • a hydroxyalkyl group typically has the formula C n H (2n+ i X) (OH) x
  • n is typically from 1 to 10, more preferably from 1 to 6, most preferably from 1 to 3.
  • x is typically from 1 to 6, more preferably from 1 to 4.
  • “Lower alkyl” as a group means unless otherwise specified, an aliphatic hydrocarbon group which may be straight or branched having 1 to 6 carbon atoms in the chain, more preferably 1 to 4 carbon atoms such as methyl, ethyl, propyl (n-propyl or isopropyl) or butyl (n-butyl, isobutyl or t- butyl).
  • the group may be a terminal group or a bridging group.
  • Patient refers to an animal, preferably a mammal, and most preferably a human.
  • Subject refers to a human or an animal.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • the group may be a terminal group or a bridging group. If the group is a terminal group, it is bonded to the remainder of the molecule through the sulfur atom.
  • the group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
  • Some of the compounds of the disclosed embodiments may exist as single stereoisomers, racemates, and/or mixtures of enantiomers and /or diastereomers. All such single stereoisomers, racemates and mixtures thereof, are intended to be within the scope of the subject matter described and claimed.
  • Formula (I) is intended to cover, where applicable, solvated as well as unsolvated forms of the compounds.
  • each formula includes compounds having the indicated structure, including the hydrated as well as the non-hydrated forms.
  • compounds of the invention may contain more than one asymmetric carbon atom.
  • the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included.
  • the use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of the invention and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present.
  • optionally substituted means the group to which this term refers may be unsubstituted, or may be substituted with one or more groups independently selected from alkyl, alkenyl, alkynyl, thioalkyl, cycloalkyl, aminocycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkylalkenyl, heterocycloalkyl, cycloalkylheteroalkyl, cycloalkyloxy, cycloalkylaminocarbonyl, cycloalkenyloxy, cycloamino, halogen, carboxyl, haloalkyl, haloalkenyl, haloalkynyl, alkynyloxy, heteroalkyl, heteroalkyloxy, hydroxyl, hydroxyalkyl, alkoxy, alkyloxyalkyloxyalkyl, cycloalkylalkyloxyalkyl,
  • the halogen is chlorine, fluorine, bromine or iodine
  • the alkyl is an optionally substituted C C n alkyl
  • the alkenyl is an optionally substituted C C n alkenyl
  • the alkynyl is a Q-Q2 alkynyl
  • the thioalkyl is an optionally substituted C 1 -Q2 thioalkyl comprising 1 or 2 sulfur atoms
  • the alkyloxy is an optionally substituted Q-Ce alkyl-O- group
  • the cycloalkyl is an optionally substituted C3-C9 cycloalkyl
  • the aminocycloalkyl is an optionally substituted C3-C9 aminocycloalkyl
  • the cycloalkylalkyl is an optionally substituted C 3 to C 9 cycloalkylalkyl
  • the cycloalkenyl is an optionally substituted C 3 -C 9 cycloalkenyl
  • the nitroalkyl is an optionally substituted Q-C 12 alkyl having at least one nitro group
  • the nitroalkenyl is an optionally substituted C C n alkenyl having at least one nitro group
  • the nitroalkynyl is an optionally substituted C C n alkynyl having at least one nitro group
  • the nitroheterocyclyl is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and having at least one nitro group
  • the optionally substituted aryl is an optionally substituted Ce-Qs aryl
  • the heteroaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S
  • alkylamino is an optionally substituted alkyl-NH- group having a C
  • alkynyl amino is an optionally substituted alkynyl-NH- group having a C C 6 alkynyl group
  • alkyloxyalkyl is an optionally substituted alkyloxy group having a C C 6 alkyl group
  • the alkyloxyalkyloxyalkyl is an optionally substituted Q to C 6 alkyloxyalkyl substituted with a Q to C 6 alkyloxyalkyl
  • cycloalkylalkyloxyalkyl is a Q to C alkyloxyalkyl substaituted with a C 3 to C 8 cycloalkyl group
  • alkyloxyaryl is an optionally substituted alkyloxy group having an optionally substituted Ce-Qs aryl
  • alkyloxycarbonyl is a an optionally substituted Q-C16 alkyloxy having a carbonyl group
  • alkyloxycyclocarbonyl is an optionally substituted optionally substituted C 3
  • pharmaceutically acceptable salts refers to salts that retain the desired biological activity of the above -identified compounds, and include pharmaceutically acceptable acid addition salts and base addition salts.
  • Suitable pharmaceutically acceptable acid addition salts of compounds of Formula (I) may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, sulfuric, and phosphoric acid.
  • Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, heterocyclic carboxylic and sulfonic classes of organic acids, examples of which are formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, fumaric, maleic, alkyl sulfonic, arylsulfonic. Additional information on pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Co., Easton, PA 1995. In the case of agents that are solids, it is understood by those skilled in the art that the inventive compounds, agents and salts may exist in different crystalline or polymorphic forms, all of which are intended to be within the scope of the present disclosure and specified formulae.
  • Prodrug means a compound that undergoes conversion to a compound of formula (I) within a biological system, usually by metabolic means (e.g. by hydrolysis, reduction or oxidation). For example an ester prodrug of a compound of formula (I) containing a hydroxyl group may be converted by hydrolysis in vivo to the parent molecule.
  • Suitable esters of compounds of formula (I) containing a hydroxyl group are for example formates, acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis- ⁇ - hydroxynaphthoates, gentisates, isethionates, di-/?-toluoyltartrates, methanesulfonates, ethanesulfonates , benzenesulfonates , p-toluenesulfonates, cyclohexylsulfamates, and quinates.
  • ester prodrug of a compound of formula (I) containing a carboxy group may be convertible by hydrolysis in vivo to the parent molecule.
  • ester prodrugs are those described by FJ. Leinweber, Drug Metab. Res., 18:379, 1987.
  • an acyl prodrug of a compound of formula (I) containing an amino group may be converted by hydrolysis in vivo to the parent molecule.
  • prodrugs for these and other functional groups, including amines are described in Prodrugs: Challenges and Rewards (Parts 1 and 2); Ed V. Stella, R. Borchardt, M. Hageman, R.Oliyai, H. Maag and J Tilley; Springer, 2007)
  • terapéuticaally effective amount or “effective amount” is an amount sufficient to effect beneficial or desired clinical results.
  • An effective amount can be administered in one or more administrations.
  • An effective amount is typically sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state.
  • the term "functional equivalent” is intended to include variants of the specific protein lysine methyl transferase species described herein. It will be understood that the protein lysine methyl transferases may have isoforms, such that while the primary, secondary, tertiary or quaternary structure of a given protein lysine methyl transferase isoform is different to the prototypical protein lysine methyl transferase, the molecule maintains biological activity as a protein lysine methyl transferase. Isoforms may arise from normal allelic variation within a population and include mutations such as amino acid substitution, deletion, addition, truncation, or duplication. Also included within the term “functional equivalent” are variants generated at the level of transcription. Enzymes have isoforms that arise from transcript variation. Other functional equivalents include protein lysine methyl transferases having altered post-translational modification such as glycosylation.
  • reprogramming cells is intended to include erasure and remodeling of epigenetic marks, such as DNA methylation, during mammalian development.
  • the term "about”, in the context of concentrations of components of the formulations, typically means ⁇ 10% of the stated value, more typically ⁇ 7.5% of the stated value, more typically ⁇ 5% of the stated value, more typically ⁇ 4% of the stated value, more typically ⁇ 3% of the stated value, more typically, ⁇ 2% of the stated value, even more typically ⁇ 1% of the stated value, and even more typically ⁇ 0.5% of the stated value.
  • range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosed ranges. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • FIG. 1 shows the effect of compounds on the methyltransferase activity of SMYD3 using MAP3K2 peptide as a substrate and refers to dose-response curves showing the effect of compounds on the methyltransferase activity of SMYD3 using MAP3K2 peptide as a substrate ; for compound A066 (Fig. 1A), for compound A088 (Fig. IB); for compound B019 (Fig. 1C); and for compound A074 (Fig. ID).
  • FIG. 2 refers to dose -response curves showing that SMYD3 compounds inhibit the proliferation of HCC, Colorectal, Lung and Pancreatic carcinoma cell lines.
  • Fig. 2A is a dose response curve showing inhibition of HepG2
  • Fig. 2B is a dose response curve showing inhibition of HCT116
  • Fig. 2C is the dose response curve showing inhibition of A549
  • Fig. 2D is the dose response curve showing inhibition of CFPAC-1
  • Fig. 2E is a dose response curve showing inhibition of HPAF-II.
  • FIG. 3 refers to an image of aWestern blot showing SMYD3 target engagement and inhibition of MAP3K2 methylation following treatment with 25.0 ⁇ of compound B019 and X4, in HEK293 cells transiently transfected with Myc-SMYD3.
  • FIG. 4 refers to colony images in 24-well plate and the corresponding dose response curves of 2 sets of experiments using HepG2 cells.
  • Fig. 4A is a colony image and
  • Fig. 4B is a dose response curve with compound A074.
  • Fig. 4C is a colony image and
  • Fig. 4D is a dose response curve with compound B019.
  • Z 1 and Z 2 may be selected from O, S or NH.
  • Z 1 may be O.
  • Z 2 may be O.
  • X may be a halogen. X may be chloro.
  • R 1 and R 2 may be independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 1 and R 2 may optionally be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
  • R 1 and R 2 may optionally form an optionally substituted aryl or optionally substituted heteroaryl together with the ring atoms that they are bond to.
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 may be independently absent, or selected from the group consisting of a bond, H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 may be independently hydrogen or methyl.
  • R 3 , R 4 , R 5 , R 6 , R 7 or R 8 may be taken together to form an optionally substituted cycloalkyl, or optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
  • Y may be selected from the group consisting of R 9 , OR 9 or NHR 9 , wherein R 9 is an optionally substituted C 3 to C 10 alkyl, optionally substituted C 3 to C 10 alkenyl, optionally substituted C 3 to C 10 alkynyl, optionally substituted C 3 to C 7 cycloalkyl, optionally substituted C 2 to Ci 0 haloalkyl, a substituted 5-membered heteroaryl comprising two or three heteroatoms selected from N, O or S or a Ci to C 2 alkyl substituted with an optionally substituted 5-membered heterocycloalkyl comprising one to two heteroatoms selected from N, O or S.
  • the compound of Formula (I) may include a pharmaceutically acceptable form or prodrug thereof.
  • the compound may have the following Formula (II):
  • the optionally substituted alkyl may be an optionally substituted Q-C 12 alkyl.
  • the optionally substituted alkyloxy may be an optionally substituted C C i6 alkyloxy.
  • the optionally substituted cycloalkyl may be an optionally substituted C3-C9 cycloalkyl.
  • the optionally substituted heterocycloalkyl may be an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S
  • the optionally substituted aryl may be an optionally substituted Ce-Qg aryl
  • the optionally substituted heteroaryl may be an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S
  • the optionally substituted alkenyl may be an optionally substituted C 2 -Q 2 alkenyl or the optionally substituted alkynyl may be an optionally substituted C 2 -Q 2 alkynyl.
  • R 9 may be a C 3 to C 10 alkyl, optionally substituted C 3 to C 6 alkenyl, optionally substituted C 2 to C 10 haloalkyl, or in each case C 3 to C 9 alkyl or C 3 to C 7 cycloalkyl, or substituted oxazolyl, isoxazolyl, 1,2-azole, pyrazolyl, triazolyl, or methylpyrrolidinonyl.
  • the compound may have the following formula (Ila):
  • a 1 may be O or NH.
  • R 10 may be a Q to C 9 alkyl or a C to C 7 cycloalkyl.
  • R 10 may be selected from the group consisting of methyl, isobutyl and cyclopropyl.
  • R 1 and R 2 may be independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted phenyl, optionally substituted pyrazolyl, optionally substituted thiazolyl, optionally substituted thiophenyl, optionally substituted benzo[d]imidazolyl, optionally substituted indolyl, optionally substituted isoindoyl, optionally substituted indazolyl, optionally substituted pyrrolyl, optionally substituted pyridinyl, optionally substituted benzyl, optionally substituted benzo[d]dioxolyl, optionally substituted benzotriazolyl, optionally substituted benzoxazolyl, optionally substituted benzofuranyl, optionally substituted pyrazolopyridinyl, optionally substituted pyrrolopyrimidinyl, optionally substituted pyrrolopyridinyl, optionally substituted naphthyrid
  • the compound may have the following Formula (lib):
  • R 1 may be H or halogen.
  • R 2 may be selected from the group consisting of H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-hydroxyphenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 2-(trifluoromethyl)phenyl, 3-(trifluoromethyl)phenyl, 4- (trifluoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4-fluoromethylphenyl, 2- hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphen
  • the compound may have the following Formula (III):
  • R 1 and R lla may be independently selected from the group consisting of H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R nb may be absent, H or optionally substituted alkyl.
  • a 2 may be selected from CH, N, O or S;
  • p may be an integer selected from 0, 1 or 2.
  • the A 2 linked group may represent R lla or R nb .
  • the A 2 linked group may represent R l la .
  • the compound may have the following Formula (Ilia):
  • R 1 and R lla may be independently selected from the group consisting of a bond, H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-hydroxyphenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 2-(trifluoromethyl)phenyl, 3-(trifluoromethyl)phenyl, 4- (trifluoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4-fluoromethylphenyl, 2- hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphenyl
  • the compound may have the following Formula (Ilia'):
  • R 1 and R lla may be independently selected from the group consisting of bond, H, cyano, methyl, ethyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-(trifluoromethyl)phenyl, 3- (trifluoromethyl)phenyl, 4-(trifiuoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl,
  • R 1 and R 2 may be taken together to form an optionally substituted 5-membered cycloalkyl, an optionally substituted 6-membered cycloalkyl, an optionally substituted 5-membered heterocycloalkyl or an optionally substituted 6-membered heterocycloalkyl.
  • the compound may have the following Formula (IV):
  • a 3 and A 4 may be independently selected from CH or N.
  • a 5 and A 6 may be independently selected from CH, N, O or S.
  • R 12a , R 13a , R 14 and R 15 may be independently selected from the group consisting of H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
  • R 12b and R 13b may be independently absent, H or an optionally substituted alkyl
  • q and r may be independently integers selected from 0, 1 or 2.
  • the A 5 linked group may represent R 12a or R 12b .
  • the A 5 linked group may represent R 12a .
  • the A 6 linked group may represent R 13a or R 13b .
  • the A 6 linked group may represent R 13a .
  • a 3 and A 4 may be both C.
  • the compound may have the following Formula (IV a):
  • R 9 may be an optionally substituted C 3 to C 10 alkyl, optionally substituted C 3 to C 10 alkenyl, optionally substituted C 3 to C 10 alkynyl or optionally substituted C 3 to C 7 cycloalkyl.
  • R 12a , R 13a , R 14 and R 15 may be independently selected from H, methyl, ethyl, propyl, butyl, halogen, cyano, COOMe, COOEt, phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-(3-methyl)pyridinyl, 2-(4- methyl)pyridinyl, 2-(5-methyl)pyridinyl, 2-(6-methyl)pyridinyl, 3-(2-methyl)pyridinyl, 3-(4- methyl)pyridinyl, 3-(5-methyl)pyridinyl, 3-(6-methyl)pyridinyl, 4-(2-methyl)pyridinyl, 4-(3- fluoro)pyridinyl, 2-(3-fluoro)pyridinyl, 2-(4-fluoro)pyridinyl, 2-(5-fluoro)pyridinyl, 2-(6- fluoro)pyr
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 may be independently selected from the group consisting of a bond, H, methyl, (S)-methyl, (R)-methyl, ethyl, (S)-ethyl, (R)-ethyl, cyano, -CH 2 OH, (S)-CH 2 OH, (R)-CH 2 OH, COOCH 3 , (S)-COOCH 3 , (R)-COOCH 3 , CH 2 OC(0)CH 3 , (R)-CH 2 OC(0)CH 3 , (S)-CH 2 OC(0)CH 3 , CH 2 OC(0)CH 2 CH 2 OCH 3 , (R)-CH 2 OC(0)CH 2 CH 2 OCH 3 , (S)-CH 2 OC(0)CH 2 CH 2 OCH 3 , (S)-CH 2 OC(0)CH 2 CH 2 OCH 3 , CH 2 CH 2 OH, (R)-CH 2 CH 2 OH, (S
  • R 4 and R 5 or R 6 and R 7 may be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
  • R 4 and R 5 or R 6 and R 7 may be taken together to form a cyclopropane.
  • R 3 and R 4 , R 3 and R 5 , R 3 and R 6 , R 3 and R 7 , R 3 and R 8 , R 4 and R 6 , R 4 and R 7 , R 4 and R 8 , R 5 and R 6 , R 5 and R 7', R' and R 8°, R 6 u and R 8 or R 7 and R 8° may be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
  • the alkylene bridge may be a 1-carbon, 2-carbon or 3-carbon alkylene bridging group wherein -CH- units may optionally be replaced with -NH-, O or S.
  • X preferably represents chlorine, bromine or fluorine.
  • R 1 and R 2 independently from another preferably represent of a bond, H, cyano, Q-C4 alkyl, in each case optionally cyano, fluorine, chlorine, bromine, amino, di(Q- C 6 alkyl ) amino, nitro, carbamoyl, Q- C 6 alkyl, Q-C 6 haloalkyl, C 3 -C 6 alkene, C 3 -C 6 alkyne, Q-C4 alkoxy, Q-C4 alkyl-CN, Q-C 4 - alkylamino, C 1 -C4 alkyl-CO-NH 2 , C 1 -C4 alkyl-NH-C(NH)-NH 2 , C 1 -C4 alkyl-C 3 -C 6 cycloalkyl, Q- C 4 alkylamino, Q-C4 alkyl- di (Ci-C 3 -alkyl
  • R 1 and R 2 preferably may optionally be taken together to form a optionally substituted C3-C5 alkylene bridge wherein one -CH 2 - unit may be replaced with -NH-, S or O and wherein the optional substituents of the alkylene bridge are selected from cyano, fluorine, chlorine, bromine, carbamoyl, Q-C 6 alkyl, Q-Q haloalkyl, C 3 - C 6 alkene, C 3 -C 6 alkyne, Q-C 4 alkoxy, C C 4 alkyl-CN, C C 4 alkylamino, Q-C 4 alkyl-CO-NH 2 , Q-C 4 alkyl-NH-C(NH)-NH 2 , Q-C 4 alkyl-C 3 -C 6 cycloalkyl, C C 4 alkylamino, Q-C4 alkyl-di(Ci-C alkyl) amino, C 1 -C4 alkyl-CO-Ci-C 4
  • R 1 and R 2 form an optionally substituted -CH 2 -CH 2 -CH 2 -CH 2 - bridge.
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are independently absent, or represent a bond, H, cyano, carbamoyl, -COO- C 1 -C 4 alkyl, -CO-NH- C 1 -C 4 alkyl, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, C 1 -C 4 alkyl-OH, C 1 -C 4 alkyl-CO- NH 2 , C 1 -C 4 alkyl-CO- C 1 -C 4 alkyl, Q- C 4 alkyl-CO- di (C C 3 alkyl) amino, C 1 -C 4 alkyl-CO- NH- Q- C 3 alkyl, d-C 4 alkyl-O-CO- C r C 4 alkyl or Q- C 4 alkyl-O-CO- C C 3 alkyl-O- Q-Q-alkyl.
  • R 3 , R 4 , R 5 , R 6 , R 7 or R 8 may also be taken together to form a Q-C 4 alkylene bridge.
  • Y preferably represents R 9 , OR 9 , or NH-R 9 wherein R 9 represents C 3 -C 6 alkyl, C 3 -C 6 alkenyl, optionally substituted C 3 -C 10 alkynyl or a in each case optionally C C 6 alkyl or C 3 -C 6 cycloalkyl substituted isoxazolyl, oxazolyl or pyrazolyl.
  • Y most preferably is OR 9 , if R 9 is C 3 -C 6 alkyl, C 3 -C 6 alkenyl, optionally substituted C 3 -C 10 alkynyl.
  • R 9 is preferably C to C 9 alkyl, optionally substituted C to Ce alkenyl or a C to C 9 alkyl, or C to C 7 cycloalkyl substituted oxazolyl, isoxazolyl or pyrazolyl.
  • R 9 most preferably is propyl.
  • R 1 is preferably methyl or H, most preferably H, may be specifically mentioned.
  • the compound may be selected from the group consisting of: ⁇
  • the compound is an enzyme inhibitor.
  • the compounds may be a protein lysine methyltransferase (PKMT) inhibitor. It may be an inhibitor for SET domain -containing and non-SET domain-containing methyl transferases.
  • the protein lysine methyltransferase may be SMYD3. SMYD3 may also methylate other substrates such as the retinoblastoma (RB I) protein or the vascular endothelial growth factor receptor 1 (VEGFR1) protein.
  • RB I retinoblastoma
  • VEGFR1 vascular endothelial growth factor receptor 1
  • the compound inhibits methylation of a histone.
  • the histone may be of the HI, H2A, H2B, H3 or H4 family.
  • the histone may be of the H1F, H1H1, H2AF, H2A1, H2A2, H2BF, H2B 1, H2B2, H3A1, H3A2, H3A3, H41 or H44 subfamily.
  • the histone may be H1F0, H1FNT, H1FOO, H1FX, HIST1H1A, HIST1H1B, HIST1H1C, HIST1H1D, HIST1H1E, HIST1H1T, H2AFB1, H2AFB2, H2AFB3, H2AFJ, H2AFV, H2AFX, H2AFY, H2AFY2, H2AFZ, HIST1H2AA, HIST1H2AB, HIST1H2AC, HIST1H2AD, HIST1H2AE, HIST1H2AG, HIST1H2AI, HIST1H2AJ, HIST1H2AK, HIST1H2AL, HIST1H2AM, HIST2H2AA3, HIST2H2AC, H2BFM, H2BFS, H2BFWT, HIST1H2BA, HIST 1H2BB, HIST 1H2BC, HIST1H2BD, HIST1H2BE,
  • the compound inhibits methylation of histone by inhibiting lysine methyltransferases.
  • the compound may inhibit ASH1L, DOT1L, EHMT1, EHMT2, EZH1, EZH2, MLL, MLL2, MLL3, MLL4, MLL5, NSD1, NSD2, NSD3, PRDM2, PRDM9, SET, SETBP1, SETD1A, SETD1B, SETD2, SETD3, SETD4, SETD5, SETD6, SETD7, SETD8, SETD9, SETDB1, SETDB2, SETMAR, SMYD1, SMYD2, SMYD3, SMYD4, SMYD5, SUV39H1, SUV39H2, SUV420H1, or SUV420H2.
  • the compound inhibits the trimethylation of histone H3 at lysine 4 (H3K4me3) and/or methylation of histone H4 at lysine 5 (H4K5me).
  • SMYD3 may regulate multiple overlapping MAP kinase pathway proteins. Accordingly, the compound of the present disclosure may modulate myostatin transcription and/or c-Met transcription.
  • the compound is assumed to inhibit the MEK-ERK mitogen-activated protein-kinase pathway.
  • the compound may inhibit methylation of a lysine residue on MAP3K2.
  • the lysine residue may be K260.
  • the compound may be administered alone or in the form of a pharmaceutical composition in combination with a pharmaceutically acceptable carrier, diluent or excipient.
  • a pharmaceutically acceptable carrier diluent or excipient.
  • the compounds while effective themselves, may be typically formulated and administered in the form of their pharmaceutically acceptable salts as these forms are typically more stable, more easily crystallized and have increased solubility.
  • the compound may, however, typically be used in the form of pharmaceutical compositions which are formulated depending on the desired mode of administration.
  • a pharmaceutical composition may comprise a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, and a pharmaceutically acceptable excipient.
  • the compositions may be prepared in manners well known in the art.
  • the amount of compound in the compositions may be such that it is effective to measurably inhibit one or both of methylation of histone H3 at lysine 4 (H3K4me3) and of histone H4 at lysine 5 (H4K5me) in a biological sample or in a patient.
  • the composition may be formulated for administration to a patient in need of such composition. In using the compounds, they may be administered in any form or mode which may make the compound bioavailable.
  • One skilled in the art of preparing formulations can readily select the proper form and mode of administration depending upon the particular characteristics of the compound selected, the condition to be treated, the stage of the condition to be treated and other relevant circumstances.
  • compositions of this disclosure may include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene -polyoxypropylene -block polymers
  • compositions as defined above may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this disclosure may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions as defined above may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions for parenteral injection may comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use.
  • suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
  • Proper fluidity may be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservative, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of micro-organisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminium monostearate and gelatin.
  • the compounds may be incorporated into slow release or targeted delivery systems such as polymer matrices, liposomes, and microspheres.
  • the injectable formulations may be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
  • compositions as defined above may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions as defined above may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations may be readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract may be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of compounds as defined above may include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutically acceptable compositions may be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers may include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylaikonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • compositions as defined above may also be administered by nasal aerosol or inhalation.
  • Such compositions may be prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • compositions as defined above may be formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions as defined above may be administered without food. In other embodiments, pharmaceutically acceptable compositions as defined above may be administered with food.
  • compositions may vary depending upon the host treated, the particular mode of administration.
  • the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present disclosure in the composition will also depend upon the particular compound in the composition.
  • a method of inhibiting SMYD3 in a cell may comprise administering to a cell a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above.
  • the activity of a compound as an inhibitor may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays may include assays that determine inhibition of either the methylation activity and/or the subsequent functional consequences, or methylation activity of one or both of histone H3 at lysine 4 (H3K4me3) and histone H4 at lysine 5 (H4K5me), or the methylation of a lysine residue on MAP3K2.
  • H3K4me3 histone H3 at lysine 4
  • H4K5me histone H4 at lysine 5
  • SMYD3 catalyzes the methylation of the MAP3K2 peptide substrate by transferring a methyl group from SAM to MAP3K2 peptide and further converts the SAM to SAH.
  • the SMYD3 methyltransferase activity is measured based on the amount of SAH produced from the reaction through the use of coupling
  • the inhibition of SMYD3 further comprises the inhibition of cell proliferation.
  • the cell may be in vitro.
  • the cell may be from a cell line.
  • the cell line may be an immortalized cell line, a genetically modified cell line or a primary cell line.
  • the cell line may be selected from the group consisting of HepG2, HCT116, A549, HPAF-II, CFPAC-1, HuH7, SNU398, Hep3B, PLC/PRF/5, HuHl, Bel7404, HCCLM3, HLE, SK-HEP-1, Mahlavu, JHH1, JHH2, JHH4, JHH5, JHH7, SNU354, SNU368, SNU387, SNU423, SNU449, SNU739, SNU761, SNU886, MIA PaCa-2 and HEK293.
  • the cell may be from tissue of a subject.
  • the cell may be in a subject.
  • a method of treating a SMYD-3-related disorder may comprise administering to a subject in need of treatment a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above.
  • the method as disclosed above may further comprise the step of administering an additional therapeutic agent in the subject.
  • the compound as disclosed above, or a pharmaceutically form or prodrug thereof, or a composition as disclosed above may be for use in therapy.
  • a compound as disclosed above or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above, may be in the manufacture of a medicament for treatment of a SMYD3-related disorder.
  • the medicament may be administered with an additional therapeutic agent, wherein said medicament may be administered in combination or alteration with the additional therapeutic agent.
  • a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above, may be for use in the treatment of a SMYD3-related disorder.
  • the disorder may be cancer, angiogenic disorder or pathological angiogenesis, fibrosis or inflammatory conditions.
  • the disorder may be lymphoma, cutaneous T-cell lymphoma, follicular lymphoma, or Hodgkin lymphoma, cervical cancer, ovarian cancer, breast cancer, lung cancer, prostate cancer, colorectal cancer, gastric cancer, pancreatic cancer, sarcoma, hepatocellular carcinoma, leukemia or myeloma, retinal angiogenic disease, liver fibrosis, kidney fibrosis, or myelofibrosis.
  • the compound may be administered with an additional therapeutic agent, wherein said medicament may be administered in combination or alteration with the additional therapeutic agent.
  • a process for synthesizing the compound as disclosed above, having the following Formula (III), may comprise the steps of:
  • R is selected from the group consisting of H, methyl, COOMe and COOEt;
  • step (b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen
  • step (c) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form the compound of formula (III);
  • step (b) and (c) may be performed simultaneously, sequentially or in any order.
  • the compounds, esters, amides, salts and solvates of Formula (III) may be prepared by a process which comprises an initial reaction step (a) between an aminobenzoate and a carbonyl-containing moiety of Formula (Va).
  • This reaction may be carried out in a solvent. It may occur in a high-boiling solvent.
  • the solvent may be selected from the group consisting of toluene, 1,4- dioxane, n-butanol, diphenyl ether, chlorobenzene, carbon tetrachloride, diethylene glycol, diglyme, hexamethylphosphoramide, o-xylene, m-xylene and p-xylene.
  • the reaction temperature may be in a range of about 100 to about 400 °C, or about 150 to about 400 °C, or about 200 to about 400 °C, or about 250 to about 400 °C, or about 300 to about 400 °C, or about 350 to about 400 °C, or about 150 to about 350 °C, or about 150 to about 300 °C, or about 150 to about 250 °C, or about 150 to about 200 °C, or about 150 to about 350 °C, or about 200 to about 300 °C, or about 250 to about 300 °C, e.g.
  • reaction solution may be diluted with a non-polar solvent.
  • the non-polar solvent may be selected from pentane, hexane, heptane, methyl t-butyl ether, petroleum ether and dichloromethane. The product may precipitate out of the solution.
  • the ensuing amino-enone may be treated with a halogenating reagent.
  • the halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride.
  • the reaction may be in a solvent. It may be in a non-polar solvent. It may be in a solvent selected from the group consisting of hexane, cyclohexane, benzene, toluene, 1,4-dioxane, chloroform, diethyl ether and dichloromethane. It may be at an elevated temperature.
  • the temperature may be in a range of about 30 to 120 °C, or about 50 to 120 °C, or about 70 to 120 °C, or about 90 to 120 °C, or about 110 to 120 °C, or about 30 to 100 °C or about 30 to 80 °C, or about 30 to 60 °C, or about 30 to 40 °C or at about 30 °C, or at about 50 °C, or at about 70 °C, or at about 90 °C, or at about 110 °C, or at about 130 °C.
  • the reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours.
  • the reaction product may be purified by aqueous work-up followed by chromatography.
  • the ester-containing starting material may be treated with a base in a solvent.
  • the base may be selected from a variety of bases including inorganic bases or nitrogen bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used.
  • the base may be lithium hydroxide.
  • the solvent mixture may contain two solvents. At least one of these solvents may be a polar solvent.
  • the solvent mixture may contain methanol.
  • the solvent mixture may contain 1,4-dioxane.
  • the solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane.
  • the reaction may be followed by an aqueous work-up under acidic conditions.
  • the pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
  • the selective functionalizing of the carboxylic acid of step (c) may be performed under peptide coupling reaction conditions known to the person skilled in the art.
  • it may involve a peptide coupling reagent selected from the group consisting of HATU, ⁇ , ⁇ '- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride.
  • It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used.
  • a solvent may be used.
  • the solvent may include polar aprotic solvents.
  • the solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane.
  • the reaction may be performed at room temperature.
  • the reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours.
  • the reaction product may be purified by aqueous work-up followed by chromatography.
  • a process for synthesizing the compound as disclosed above, having the following Formula (III), wherein R 1 is optionall a halogen or hydrogen, may comprise the steps of:
  • step (b) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form an amide;
  • step (c) selectively functionalizing at least one halogen of the halogenated cyclized product of step (a) with a group having the following formula (Vila) or formula (Vllb) under reaction conditions to form the compound of formula (III);
  • step (b) and (c) may be performed simultaneously, sequentially or in any order.
  • the compounds, esters, amides, salts and solvates of Formula (III) may alternatively be prepared by a process which comprises an initial reaction step (a) between an aminobenzoate and a carbonyl-containing moiety of Formula (Vb).
  • the starting materials may be treated with a halogenating reagent.
  • the halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride.
  • the reaction may occur without a solvent.
  • the temperature for mixing the solvents may be in a range of about -30 to 10 °C, or about - 20 to 10 °C, or about 10 to 10 °C, or about 0 to 10 °C, or about -30 to 0 °C, or about -30 to -10 °C or about -30 to -20 °C, or at about -30 °C, or at about -20 °C, or at about -10 °C, or at about 0 °C, or at about 10 °C.
  • the reaction temperature may be raised to about 60 to 150 °C, or to about 80 to 150°C, or to about 100 to 150°C, or to about 120 to 150°C, or to about 140 to 150°C, or to about 60 to 130°C, or to about 60 to 110°C, or to about 60 to 90°C, or to about 60 to 70°C, or to about 60°C, or to about 80°C, or to about 100°C, or to about 110°C, or to about 130°C, or to about 150°C.
  • the reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours.
  • the reaction product may be purified by aqueous work-up followed by chromatography.
  • the ester -containing starting material may be treated with a base in a solvent.
  • the base may be selected from a variety of bases including inorganic bases or nitrogen bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used.
  • the base may be lithium hydroxide.
  • the solvent mixture may contain of two solvents. At least one of these solvents may be a polar solvent.
  • the solvent mixture may contain methanol.
  • the solvent mixture may contain 1,4-dioxane.
  • the solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane.
  • the reaction may be followed by an aqueous work-up under acidic conditions.
  • the pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
  • the selective functionalizing of the carboxylic acid in step (b) may be performed under peptide coupling reaction conditions known to the person skilled in the art.
  • it may involve a peptide coupling reagent selected from the group consisting of HATU, ⁇ , ⁇ '- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride.
  • It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used.
  • a solvent may be used.
  • the solvent may include polar aprotic solvents.
  • the solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane.
  • the reaction may be performed at room temperature.
  • the reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours.
  • the reaction product may be purified by aqueous work -up followed by chromatography.
  • Step (c) may be performed under cross coupling reaction conditions known to the person skilled in the art.
  • the catalyst may involve a cross coupling catalyst.
  • the catalyst may be selected from a palladium-containing catalyst. It may be selected from the group consisting of Pd(PPh 3 ) 4 or 1, 1 '- [bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with dichlorome thane. It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used.
  • a solvent mixture may be used. The solvent mixture may contain water.
  • the solvent mixture may contain a polar solvent.
  • the solvent mixture may be a mixture of water and 1,4-dioxane.
  • the reaction temperature may be in the range of about 60 to 150 °C, or to about 80 to 150 °C, or to about 100 to 150 °C, or to about 120 to 150 °C, or to about 140 to 150 °C, or to about 60 to 130 °C, or to about 60 to 110 °C, or to about 60 to 90 °C, or to about 60 to 70 °C, or to about 60 °C, or to about 80 °C, or to about 100 °C, or to about 110 °C, or to about 130 °C, or to about 150 °C.
  • the reaction may be performed under conditions known to the person skilled in the art.
  • the reaction product may be purified by filtration followed by chromatography.
  • a process for synthesizing the compound as disclosed above having the following formula (IV), comprises the ste s of;
  • step (b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen
  • step (c) optionally selectively hydrolyzing the ester of step (a) to a carboxylic acid; and (d) selectively functionalizing the carboxylic acid of the cyclized product of step (a) or step (c) with a group having the following formula (VI) under reaction conditions to form the compound of formula (IV);
  • step (b), (c) and (d) may be performed simultaneously, sequentially or in any order.
  • reaction steps may be described as disclosed above.
  • the process comprises the step of optionally hydrolyzing the carboxylic acid ester after formation of the cyclized product in step (a).
  • the hydrolyzing step may be performed under conditions known to the person skilled in the art.
  • the compounds, esters, amides, salts and solvates of Formula (IV) may be prepared by a process which comprises an initial reaction step (a) between a terephthalic acid or an ester therof, and a carbonyl-containing moiety of Formula (VIII). This reaction may be carried out in a solvent. It may occur in a high-boiling solvent.
  • the solvent may be selected from the group consisting of toluene, 1,4-dioxane, n-butanol, diphenyl ether, chlorobenzene, carbon tetrachloride, diethylene glycol, diglyme, hexamethylphosphoramide, o-xylene, m-xylene and p-xylene.
  • the reaction temperature may be in a range of about 100 to about 400 °C, or about 150 to about 400 °C, or about 200 to about 400 °C, or about 250 to about 400 °C, or about 300 to about 400 °C, or about 350 to about 400 °C, or about 150 to about 350 °C, or about 150 to about 300 °C, or about 150 to about 250 °C, or about 150 to about 200 °C, or about 150 to about 350 °C, or about 200 to about 300 °C, or about 250 to about 300 °C, e.g.
  • reaction solution may be diluted with a non-polar solvent.
  • the non-polar solvent may be selected from pentane, hexane, heptane, methyl t-butyl ether, petroleum ether and dichloromethane. The product may precipitate out of the solution.
  • the ensuing amino-enone may be treated with a halogenating reagent.
  • the halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride.
  • the reaction may be in a solvent. It may be in a non-polar solvent. It may be in a solvent selected from the group consisting of hexane, cyclohexane, benzene, toluene, 1,4-dioxane, chloroform, diethyl ether and dichloromethane. It may be at an elevated temperature.
  • the temperature may be in a range of about 30 to 120 °C, or about 50 to 120 °C, or about 70 to 120 °C, or about 90 to 120 °C, or about 110 to 120 °C, or about 30 to 100 °C or about 30 to 80 °C, or about 30 to 60 °C, or about 30 to 40 °C or at about 30 °C, or at about 50 °C, or at about 70 °C, or at about 90 °C, or at about 110 °C, or at about 130 °C.
  • the reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours.
  • the reaction product may be purified by aqueous work-up followed by chromatography.
  • the ester-containing starting material may be treated with a base in a solvent.
  • the base may be selected from a variety of bases including inorganic bases or nitrogen bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used.
  • the base may be lithium hydroxide.
  • the solvent mixture may contain of two solvents. At least one of these solvents may be a polar solvent.
  • the solvent mixture may contain methanol.
  • the solvent mixture may contain 1,4-dioxane.
  • the solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane.
  • the reaction may be followed by an aqueous work-up under acidic conditions.
  • the pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
  • the selective functionalizing of the carboxylic acid of step (d) may be performed under peptide coupling reaction conditions known to the person skilled in the art.
  • it may involve a peptide coupling reagent selected from the group consisting of HATU, ⁇ , ⁇ '- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride.
  • It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases.
  • triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used.
  • a solvent may be used.
  • the solvent may include polar aprotic solvents.
  • the solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane.
  • the reaction may be performed at room temperature.
  • the reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours.
  • the reaction product may be purified by aqueous work -up followed by chromatography.
  • the term "at least one ketone” refers to 1, 2 or 3 ketone moieties and the term “at least one halogen” refers to 1, 2 or 3 halogen moieties.
  • a SMYD3 enzymatic assay was developed using Promega's Methyltransferase -GloTM reagents.
  • SMYD3 catalyzes the methylation of the MAP3K2 peptide substrate by transferring a methyl group from SAM to MAP3K2 peptide and further converts the SAM to SAH.
  • the SMYD3 methyltransferase activity is measured based on the amount of SAH produced from the reaction through the use of coupling enzymes that convert the SAH to ATP.
  • the MTase-Glo detection solution then catalyzes the formation of light from ATP.
  • the compounds were incubated with 0.4 ⁇ of SMYD3 enzyme for 30 min in low volume 384 well plates. A final concentration of 1.0 ⁇ and 10 ⁇ SAM and MAP3K2 peptide were added and further incubated for 30 min at room temperature before adding the MTase Glo and detection reagent. Reaction signals were detected using microplate readers on luminescent mode (Safire Tecan). The IC 50 was determined by non-linear regression, using GraphPad Prism version, 5.03.
  • the cell proliferation assays were tested in several cell lines, including HepG2, HCT116, A549, HPAF-II, CFPAC-1, HuH7, SNU398, Hep3B, and HEK293. All cell lines are from ATCC. HepG2, HPAF-II and HEK293 are cultured in Eagle's MEM media supplemented with fetal bovine serum. HCT116 is cultured in McCoy media supplemented with fetal bovine serum. Huh-7 is cultured in DMEM low glucose (1000 mg/L glucose) with 10% FBS, 1% L-Glutamate and 1% Penicillin/ Streptomycin.
  • SNU398 is cultured in RPMI with 10% FBS with 1% L-Glutamate and 1% Penicillin/ Streptomycin.
  • Hep3B is cultured in Eagle's MEM with 10% FBS, 1% L- Glutamate and 1% Penicillin/ Streptomycin.
  • A549 is cultured in RPMI media supplemented with fetal bovine serum while CFPAC-1 is cultured in IMDM media supplemented with fetal bovine serum. All media and serum are purchased from Gibco (Lifetech). All cells were grown in a temperature controlled incubator at 37 °C and 5% C0 2 .
  • Cell proliferation assay was performed using CellTiter-Glo Luminescent Cell Viability Assay (Promega) following manufacturer's instructions.
  • the cell -line of interest was treated with compounds that were serial diluted in its respective media. Plates were incubated for 3 days at 37 °C in 5% C0 2 . After 3 days, an equal volume of Cell Titer Glo reagent was added. Plates were rocked on a rotator for 2 h. 100 ⁇ L ⁇ of each well were transferred to a 96-well opaque plate, and luminescence emitted was measured with the Tecan Safire II.
  • the target engagement assay was performed with HEK293 that is engineered to overexpress SMYD3 (HEK293-SMYD3).
  • the plasmid SMYD3 (Myc-DDK-tagged-Human SET and MYND domain containing 3) was purchased from Origene (RC230064) and transfected with lipofectamine 2000 (Invitrogen) into HEK293 (ATCC).
  • the cell line is cultured in Eagle's MEM media supplemented with fetal bovine serum and geneticin (Invitrogen). Presence of over -expressed SMYD3 is confirmed with western blot with antibody against SMYD3 and the MYC tag as well as with RT-PCR with SMYD3 primers.
  • the cells were seeded in 6 well plates. After seeding for 24 h, the cells were treated with either DMSO or 25 ⁇ compound and incubated for 24hr. The cells were trypsinized and the lysate was extracted with RIPA buffer (Santa Cruz). The total protein concentration of lysate is quantified using the standard Bradford assay (Biorad protein assay, microplate standard assay).
  • Hep G2 cells were purchased from ATCC. Hep G2 cells were maintained in Eagle's Minimum Essential Medium (Sigma, Cat No: #M0643) supplemented with 10% Fetal Bovine Serum (Hyclone, Cat No: SV30087.03), 2 mM L-glutamine, 100 units/mL penicillin and 100 ⁇ g/mL streptomycin (Life Technologies, Cat No: 10378-016). The soft agar assays were performed in concordance to the ETC approved method report for soft agar assay (ETC document number: RD0019). Briefly, 600 ⁇ of 0.6% agar was added to 24-well plate (Corning, Cat No: 3738) to form the base layer.
  • ETC document number: RD0019 ETC document number: RD0019
  • SMYD3 compounds inhibit the proliferation of different cancer cells
  • SMYD3 compounds inhibit the SMYD3 mediated methylation of MAP3K2 in cells
  • SMYD3 target engagement with B019 was demonstrated in HEK293 cells transiently transfected with Myc-tagged SMYD3.
  • the cells were treated with 25 ⁇ of the compound, overnight before analyzing the lysate on Western Blot.
  • a 35-37% reduction in SMYD3 levels was observed using an antibody (Anti-SMYD3 #Abl77163) (1 :5000) against SMYD3, as shown in Fig. 3.
  • B019 was further tested for its ability to inhibit SMYD3 in cells through its effects on cellular MAP3K2 methylation. This was performed using an antibody against the MAP3K2 (me2/me3), Anti- ME2/ME3-K260-MAP3K2 (1 :500) and total MAP3K2, Anti-MEKK2 #ab33918 (1: 10,000). B019 inhibited the methylation of MAP3K2 in the cells without changing the total MAP3K2 levels. Whereas, the less active compound X4 (please refer to Comparative Example 1) was unable to inhibit the methylation of MAP3K2 at 25 ⁇ .
  • Step 2 General Procedure B Cyclized product SI (1 equiv) was treated with phosphorus oxychloride (10 mL/g) and the mixture was heated at 100 °C for 4 h. Upon cooling, the crude mixture was either: (a) concentrated under reduced pressure, diluted with cold water and stirred until a free solid formed. The solid was collected by filtration and washed with hexanes to afford compound S2, or: (b) diluted with dichloromethane and poured into a separating funnel containing cold 2 M aqueous sodium hydroxide. The organic layer was separated and the aqueous layer was extracted thrice with dichloromethane.
  • Step 3 see General Procedure C for synthesis ofS4.
  • Step 4 see General Procedure C to synthesize compounds with general formula (III). 5.
  • Step 1 Synthesis of intermediate S8.
  • Step 3 Synthesis of intermediate S10, See General Procedure C.
  • Table 3 shows a list representing the exemplified compounds of this disclosure, together with the biological activity data.
  • the ability of the exemplified compounds to inhibit the catalytic function of SMYD3 was tested using the MTase assay by using the MAP3K2 as a peptide substrate. The compounds were found to inhibit the methyltransferase activity of SMYD3.
  • Compound A001 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl piperazine- 1-carboxylate as starting materials.
  • A002 Compound A002 was prepared according to General Procedure CI, step 3a, using commercially available 9-chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and allyl piperazine-l-carboxylate as starting materials.
  • Compound A003 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and valeric acid as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,8-diazabicyclo[3.2.1]octane-3- carboxylate to give teri-butyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8- diazabicyclo[3.2.1]octane-3-carboxylate.
  • Step 2 The above product (190 mg, 0.417 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (8 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,8-diazabicyclo[3.2.1]octan- 8 -yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
  • Step 3 The crude material from above (72 mg, 0.202 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (56 ⁇ L ⁇ , 0.405 mmol, 2 equiv) followed by n-propyl chloroformate (34 ⁇ , 0.303 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A004 as a white solid (10.0 mg, 11%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,5-diazabicyclo[2.2.2]octane-2- carboxylate to afford teri-butyl 5-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5- diazabicyclo[2.2.2]octane-2-carboxylate.
  • Step 2 The above intermediate (120 mg, 0.263 mmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (8 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (2,5-diazabicyclo[2.2.2]octan- 2-yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
  • Step 3 The crude material (70 mg, 0.197 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (39.8 mg, 0.393 mmol, 2 equiv) followed by n-propyl chloroformate (36.2 mg, 0.295 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A005 as a white solid (10.0 mg, 11%) upon lyophilization.
  • Compound A006 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-cyclopropylisoxazole-3-carboxylic acid as starting materials.
  • Compound A007 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-isobutylisoxazole-3-carboxylic acid as starting materials.
  • Compound A008 was prepared according to General Procedure A, B and C2 using 4- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,8-diazabicyclo[3.2.1]octane-8- carboxylate to give teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate.
  • Step 2 The resulting intermediate (220 mg, 0.482 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,8-diazabicyclo[3.2.1]octan- 3 -yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
  • Step 3 The crude material from above (190 mg, 0.534 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.09 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 0.82 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A009 as a white solid (100 mg, 42%) upon lyophilization.
  • Compound A010 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and (S)-n-propyl 3-methylpiperazine-l- carboxylate as starting materials.
  • Compound A011 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and (R)-n-propyl 3-methylpiperazine-l- carboxylate as starting materials.
  • Compound A012 was prepared according to General Procedure CI using (9-chloro-5, 6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-methylisoxazole-3-carboxylic acid as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 7, 9-diazabicyclo[3.3.1]nonane-9- carboxylate to afford teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9- diazabicyclo[3.3.1]nonane-9-carboxylate.
  • Step 2 The resulting intermediate (230 mg, 0.489 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (3,9-diazabicyclo[3.3.1]nonan-3- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A013 as a white solid (50 mg, 41%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,6-diazabicyclo[3.1.1]heptane-3- carboxylate to give teri-butyl 6-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6- diazabicyclo[3.1.1 ]heptane-3-carboxylate
  • Step 2 The resulting intermediate (200 mg, 0.453 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (3,6-diazabicyclo[3.1.1]heptan-6- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.293 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.99 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A014 as a white solid (50 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,9-diazabicyclo[3.3.1]nonane-3- carboxylate to afford teri-butyl 9-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9- diazabicyclo [3.3.1 ] nonane-3 -carboxylate.
  • Step 2 The resulting intermediate (200 mg, 0.426 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,9-diazabicyclo[3.3.1]nonan- 9-yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
  • Step 3 The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A015 as a white solid (50 mg, 41%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-ethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-ethylpiperazine-l-carboxylate
  • Step 2 The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (2-ethylpiperazin- 1 -yl)methanone .
  • Step 3 The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A016 as a white solid (50 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,5-dimethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5-dimethylpiperazine-l- carboxylate
  • Step 2 The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,5-dimethylpiperazin- 1 -yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A017 as a white solid (50 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-methylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-methylpiperazine-l-carboxylate.
  • Step 2 The resulting intermediate (160 mg, 0.36 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(3-methylpiperazin-l-yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.291 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.00 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.50 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A018 as a white solid (50 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl-3,6-diazabicyclo[3.1.1]heptane-6- carboxylate to give teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6- diazabicyclo[3.1.1]heptane-6-carboxylate.
  • Step 2 The resulting intermediate (160 mg, 0.362 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.to afford (3,6-diazabicyclo[3.1.1]heptan-3- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.293 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.99 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.49 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A019 as a white solid (50 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,6-dimethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,6-dimethylpiperazine-l- carboxylate.
  • Step 2 The resulting intermediate (160 mg, 0.349 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h.
  • Step 3 The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A020 as a white solid (20 mg, 16%) upon lyophilization.
  • Compound A022 was prepared according to General Procedure A, B and C2 using 3- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A023 was prepared according to General Procedure A, B and C2 using 3- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A024 was prepared according to General Procedure A, B, and C2 using 3- phenylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A025 was prepared according to the General Procedure A, B, and C2 using 2-(3- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A026 was prepared according to General Procedure A, B and C2 using 3- oxocyclohexanecarbonitrile (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
  • A027 Compound A027 was prepared according to General Procedure A, B, and C2 using 3-(pyridin-3- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A028 was prepared from A025 as starting material following General Procedure D.
  • Compound A029 was prepared according to General Procedure A, B and C2 using methyl 4- oxocyclohexanecarboxylate (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-ethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-ethylpiperazine-l-carboxylate.
  • Step 2 The resulting intermediate (220 mg, 0.48 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (3 -ethylpiperazin- 1 -yl)methanone .
  • Step 3 The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A030 as a white solid (50 mg, 34%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-ethylpiperazine-l-carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-methylpiperazine-l- carboxylate.
  • Step 2 The resulting intermediate (220 mg, 0.496 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (2-methylpiperazin- 1 -yl)methanone .
  • Step 3 The crude material from above (120 mg, 0.349 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.25 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A031 as a white solid (60 mg, 40%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl-2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
  • Step 2 The resulting intermediate (170 mg, 0.37 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (3 -(hydroxymethyl)piper azin- 1 -yl)methanone .
  • Step 3 The crude material from above (120 mg, 0.333 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.74 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.31 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A032 as a white solid (20 mg, 13%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,3-dimethylpiperazine-l-carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,3-dimethylpiperazine-l - carboxylate.
  • Step 2 The resulting intermediate (170 mg, 0.371 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,3-dimethylpiperazin- 1 -yl)methanone.
  • Step 3 The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A033 as a white solid (30 mg, 20%) upon lyophilization.
  • Compound A034 was prepared according to General Procedure A, B and C2 using methyl 2-(4- oxocyclohexyl)acetate (General Procedure A) and n-propyl piperazine-1 -carboxylate (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,2-dimethylpiperazine-l -carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,2-dimethylpiperazine-l - carboxylate.
  • Step 2 The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(3,3-dimethylpiperazin-l-yl)methanone trifluoroacetate salt.
  • Step 3 The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A035 (3%) as a yellow oil.
  • Step 1 According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 4,7-diazaspiro[2.5]octane-4- carboxylate to afford teri-butyl 7-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7- diazaspiro[2.5]octane-4-carboxylate.
  • Step 2 The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(4,7-diazaspiro[2.5]octan-7-yl)methanone trifluoroacetate salt.
  • Step 3 The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess).
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 4,7-diazaspiro[2.5]octane-7- carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7- diazaspiro[2.5]octane-7-carboxylate.
  • Step 2 The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(4,7-diazaspiro[2.5]octan-4-yl)methanone trifluoroacetate salt.
  • Step 3 The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A037 (4%) as a yellow oil.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl methyl piperazine-l,2-dicarboxylate to afford l-(teri-butyl) 2-methyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l,2- dicarboxylate.
  • Step 2 To a solution of the the above intermediate (184.2 mg, 0.378 mmol) in dichloromethane (1.1 mL) was added trifluoroacetic acid (0.59 mL, 7.71 mmol, 20 equiv). The resulting mixture was stirred for 2 h before concentrating under reduced pressure to give methyl 4-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)piperazine-2-carboxylate.
  • Step 3 The crude material from above (104 mg, 0.268 mmol) was dissolved in dichloromethane (1.5 mL) and triethylamine (0.080 mL, 0.574 mmol, 2.15 equiv) and propyl chloroformate (0.050 mL, 0.445 mmol, 1.67 equiv) were added. The mixture was stirred for 1 h before quenching by the addition of saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A042 as a yellow solid (88.7 mg, 70%) upon lyophilization.
  • Step 1 According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,5-dimethylpiperazine-l -carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,5-dimethylpiperazine-l - carboxylate.
  • Step 2 The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,6-dimethylpiperazin- 1 -yl)methanone.
  • Step 3 The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A043 as a white solid (50 mg, 34%) upon lyophilization.
  • Compound A044 was prepared according to General Procedure A, B, and C2 using methyl 3- oxocyclohexanecarboxylate (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A045 was prepared using A046 as starting material according to General Procedure D.
  • Compound A046 was prepared according to General Procedure A, B and C2 using 4- oxocyclohexanecarbonitrile (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
  • A047 Compound A047 was prepared according to General Procedure A, B, and C2 using 3- allylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Step 1 To a solution of Compound A042 (300.5 mg, 0.634 mmol) in methanol (2 mL), 1,4-dioxane (1 mL) and water (2 mL) was added lithium hydroxide (34.4 mg, 1.436 mmol, 2.26 equiv). Upon completion, ethyl acetate was added and the pH was adjusted to 3 using concentrated hydrochloric acid.
  • Step 2 To a solution of the crude intermediate (150 mg, 0.326 mmol) in dichloromethane (2 mL) and N,N-dimethylformamide (5 ⁇ ) was added oxalyl chloride (0.2 mL, 0.489 mmol, 1.5 equiv). After 2 h, the solvent was removed under reduced pressure. The residue (77.8 mg, 0.163 mmol) was re- dissolved in tetrahydrofuran (1 mL) and triethylamine (32.9 mg, 0.325 mmol, 2 equiv) followed by methylamine (5.05 mg, 0.163 mmol, 1 equiv) were added.
  • Step 1 General Procedure CI was performed between commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid and teri-butyl piperazine-l-carboxylate as starting materials to obtain teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate.
  • Step 2 The resulting intermediate (800 mg, 1.861 mmol) was dissolved in trifluoroacetic acid (2 mL) and dichloromethane (15 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (piper azin- 1 -yl)methanone .
  • Step 3 Propylamine (11 ⁇ L, 0.14 mmol, 1 equiv), N,N-diisoproylethylamine (0.1 mL, 0.57 mmol, 4.07 equiv) and N,N'-disuccinimidyl carbonate (56.3 mg, 0.21 mmol, 1.5 equiv) were dissolved in dichloromethane (1 mL). After 2 h, a solution of intermediate (46.2 mg, 0.14 mmol) and N,N- diisoproylethylamine (0.15 mL, 0.86 mmol, 6.15 equiv) in dichloromethane (2 mL) was added. After 1 h of stirring, the mixture was concentrated and purified by column chromatography (methanol/dichloromethane) to afford A049 as a white solid (30 mg, 52%) upon lyophilization.
  • Compound A051 was performed according to General Procedure CI between commercially available 9-chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and 3-methyl-l-propylpiperazine-l,3-di carboxylate as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
  • Step 2 Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (1.22 mg, 0.010 mmol, 0.05 equiv) was dissolved in dichloromethane (1 mL) at 0 °C. Triethylamine (83 ⁇ L ⁇ , 0.60 mmol, 3 equiv) followed by acetic anhydride (22 ⁇ L ⁇ , 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL).
  • Step 3 The resulting crude intermediate (80 mg, 0.159 mmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl acetate.
  • Step 4 The crude material from above (70 mg, 0.174 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 3.34 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 2.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A052 as a white solid (10 mg, 12%) upon lyophilization.
  • Compound A053 was prepared according to General Procedure E, F, C2 and G using 3-(pyridin-2- yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
  • Step 2 Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (1.22 mg, 0.010 mmol, 0.05 equiv) was dissolved in dichloromethane (1 mL) at 0 °C. Triethylamine (83 ⁇ L ⁇ , 0.60 mmol, 3 equiv) followed by isobutyryl chloride (25.6 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL).
  • Step 3 The resulting crude intermediate (80 mg, 0.151 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h.
  • Step 4 The crude material from above (60 mg, 0.14 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 4.16 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.12 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A054 as a white solid (15 mg, 21%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
  • Step 2 Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (4.9 mg, 0.040 mmol, 0.2 equiv) was dissolved in dichloromethane (5 mL) at 0 °C. Triethylamine (42 ⁇ L ⁇ , 0.30 mmol, 1.5 equiv) followed by 3-methoxypropionyl chloride (29.4 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL).
  • Step 3 The resulting crude intermediate (80 mg, 0.147 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl 3-methoxypropanoate.
  • Step 4 The crude material from above (50 mg, 0.112 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 5.19 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.89 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A055 as a white solid (8 mg, 13%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-carbamoylpiperazine-l-carboxylate to give teri-butyl 2-carbamoyl-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate.
  • Step 2 The resulting crude intermediate (98.1 mg, 0.207 mmol) was dissolved in trifluoro acetic acid (0.16 mL) and dichloromethane (0.16 mL) for 20 min. The mixture was concentrated under reduced pressure and then diluted with ethyl acetate. The organic layer was washed with saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-2-carboxarnide.
  • Step 3 The crude material from above (57.8 mg, 0.155 mmol) was dissolved in dichloromethane (1.6 mL) and triethylamine (29 mg, 0.287 mmol, 1.84 equiv) followed by n-propyl chloroformate (24.0 mg, 0.196 mmol, 1.26 equiv) were added at room temperature. The mixture was stirred for 20 min before quenching by the addition of saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (100% ethyl acetate) followed by preparative HPLC to afford A056 as a white solid (24.6 mg, 35%) upon lyophilization.
  • Compound A057 was prepared according to General Procedure A, B and C2 using 3-(pyrimidin-5- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A058 was prepared according to General Procedure A, B and C2 using 3-(pyridin-4- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • A059 Compound A059 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl 3-carbamoylpiperazine-l- carboxylate as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(2-hydroxyethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2- hydroxyethyl)piperazine- 1 -carboxylate.
  • Step 2 The resulting crude intermediate (54.3 mg, 0.115 mmol) was dissolved in trifluoroacetic acid (1.05 mL) and dichloromethane (1.5 mL) for 30 min. The mixture was concentrated under reduced pressure and then diluted with ethyl acetate. The organic layer was washed with saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3-yl)(3-(2-hydroxyethyl)piperazin-l-yl)methanone.
  • Step 3 The crude material from above (16.5 mg, 0.044 mmol) was dissolved in dichloromethane (1 mL) and triethylamine (10.88 mg, 0.108 mmol, 2.44 equiv) followed by n-propyl chloroformate (8.1 mg, 0.066 mmol, 1.5 equiv) were added at room temperature. After 20 min, saturated ammonium chloride was added and the aqueous layer was extracted with dichloromethane. The organic layers were washed with brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A060 as a white solid (4.88 mg, 24%) upon lyophilization.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
  • Step 2 Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (4.9 mg, 0.040 mmol, 0.2 equiv) was dissolved in dichloromethane (5 mL) at 0 °C. Triethylamine (30.4 ⁇ L ⁇ , 0.30 mmol, 1.5 equiv) followed by butyryl chloride (25.6 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL).
  • Step 3 The resulting crude intermediate (80 mg, 0.151 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl butyrate.
  • Step 4 The crude material from above (50 mg, 0.116 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 5 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.75 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A061 as a white solid (8 mg, 13%) upon lyophilization.
  • Step 1 According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-trifluoromethylpiperazine-l - carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- (trifluoromethyl)piperazine- 1 -carboxylate.
  • Step 2 The resulting intermediate (20 mg, 0.40 mmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2-(trifluoromethyl)piperazin-l -yl)methanone.
  • Step 3 The crude material from above (12 mg, 0.030 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 19.3 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 14.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (30 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A064 as a white solid (5 mg, 34%) upon lyophilization.
  • Compound A065 was prepared according to General Procedure A, B and C2 using 2-(3- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A066 was prepared using A078 as starting material according to General Procedure D.
  • Compound A067 was prepared was prepared according to General Procedure A, B, C2 and D using 2- (3-oxocyclohexyl)acetamide (General Procedure A) and (5) -propyl 3-methylpiperazine-l-carboxylate (General Procedure C2) as starting materials.
  • Compound A068 was prepared according to General Procedure A, B and C2 using 4- ((dimethylamino)methyl)cyclohexanone (General Procedure A) and n-propyl piperazine-1- carboxylate (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(2-methoxy-2-oxoethyl)piperazine- 1-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2-methoxy-2- oxoethyl)piperazine-l-carboxylate.
  • Step 2 The resulting intermediate (251 mg, 0.50 mmol) was dissolved in trifluoroacetic acid (0.77 mL) and dichloromethane (1.2 mL) for 30 min. The mixture was concentrated and ethyl acetate was added. The organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give methyl 2-(4-(9-chloro- 5,6,7, 8-tetrahydroacridine-3-carbonyl)piperazin-2-yl)acetate.
  • Step 3 The crude material from above (201 mg, 0.500 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (101.22 mg, 1.00 mmol, 2 equiv) followed by n-propyl chloroformate (91.4 mg, 0.75 mmol, 1.5 equiv) were added at 0 °C. After 30 min, the mixture was quenched with saturated ammonium chloride and the organic layer was separated. The aqueous layer was extracted with dichloromethane and the combined organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A069 as a white solid (208 mg, 85%) upon lyophilization.
  • Compound A070 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl 3-(2-methoxy-2- oxoethyl)piperazine-l-carboxylate as starting materials.
  • Step 1 To a solution of A070 (153.4 mg, 0.3144 mmol) in methanol (1 mL) and 1,4-dioxane (0.5 mL) was added an aqueous solution of lithium hydroxide (75.3 mg, 3.144 mmol, 10 equiv) in water (1 mL) at 0 °C. The reaction was allowed to stir for 30 min before quenching by the addition of concentrated hydrochloric acid and ethyl acetate to pH 2. The organic layer was separated and the aqueous layer was extracted thrice with ethyl acetate.
  • Step 2 To a solution of crude intermediate (106.1 mg, 0.2239 mmol) in dichloromethane (2.2 mL) and N,N-dimethylformamide (0.004 mL) was added oxalyl chloride (0.04 mL, 0.466 mmol, 2.08 equiv). When bubbling has ceased (10 min), the suspension was sonicated for 30 min and then stirred for another 1.5 h. The contents were concentrated under reduced pressure.
  • Compound A075 was prepared according to General Procedure A, B, C2 using 4-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and n-propyl piperazine- 1-carboxylate (General Procedure C2) as starting materials.
  • Compound A076 was prepared using A065 as starting material according to General Procedure D.
  • Compound A077 was prepared using A065 as starting material in dimethylsulfoxide at 0°C and potassium carbonate followed by 30% hydrogen peroxide was added. The reaction mixture was stirred at room temperature for 12 h. The reaction mass was diluted with ethyl acetate and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate and concentrated to afford A077.
  • Compound A078 was prepared according to General Procedure A, B and C2 using 2-(4- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate for Step 3 (General Procedure C2) as starting materials.
  • Step 1 According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 6,8-diazabicyclo[3.2.2]nonane-6- carboxylate to give teri-butyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-6,8- diazabicyclo[3.2.2]nonane-6-carboxylate.
  • Step 2 The resulting intermediate (300 mg, 0.638 mmol) was dissolved in trifluoroacetic acid (1.2 mL) and dichloromethane (6 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (6,8- diazabicyclo[3.2.2]nonan-6-yl)(9-chloro-5,6,7,8-tetrahydroacridin-3-yl)methanone.
  • Step 3 The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A080 as a white solid (30 mg, 24%) upon lyophilization.
  • Compound A081 was prepared according to General Procedure A, B, C2 and D using 2-(4- oxocyclohexyl)acetonitrile (General Procedure A) and (S)-n-propyl 3 -methylpiperazine- 1-carboxylate (General Procedure C2) as starting materials.
  • Compound A082 was prepared according to General procedures steps A, B and C2 using 4-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (S)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
  • Step 1 5-methylisoxazole-3-carboxylic acid (20 mg, 0.15 mmol) and teri-butyl (5)-2- methylpiperazine-l-carboxylate (40.1 mg, 0.20 mmol, 1.33 equiv) were dissolved in N,N- dimethylformamide (10 mL) before HATU (190 mg, 0.5 mmol, 3.33 equiv), N,N- diisopropylethylamine (0.35 mL, 2.0 mmol, 13.3 equiv) and 4-dimethylaminopyridine (1 mg) were added. The mixture was stirred for 4 h at room temperature before adding brine and extracting with ethyl acetate.
  • Step 2 The crude material was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h before concentrating. The crude material was purified by column chromatography (ethyl acetate/hexanes) to give (5)-(5-methylisoxazol-3-yl)(3-methylpiperazin-l-yl)methanone (15 mg, 48% over 2 steps).
  • Step 3 The intermediate from above was subjected to General Procedure CI with 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid to afford A083.
  • Compound A088 was prepared was prepared according to General procedure steps A, B, C2 and D using 2-(3-oxocyclohexyl)acetamide (General Procedure A) and (S)-n-propyl 3-methylpiperazine-l - carboxylate (General Procedure C2) as starting materials.
  • Compound A089 was prepared according to General Procedure A, B and C2 using 4-((5-methyl- l,2,4-oxadiazol-3-yl)methyl)cyclohexanone (General Procedure A) and n-propyl piperazine-1- carboxylate (General Procedure C2) as starting materials.
  • A094 Compound A094 was prepared according to General Procedure E, F, C2 and G using 3-(5- fluoropyridin-2-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.

Abstract

Compounds For Inhibition Of Cancer and Epigenesis The present invention relates to quinolines and 5,6,7,8-tetrahydroacridines of the formula (I) wherein Z1, Z2, X, R1 to R8 and Y are defined as described in the specification, or a pharmaceutically acceptable form or prodrug thereof, that are inhibitors of methyl transferases such as protein lysine methyltransferases and more particularly SMYD3. The present invention also relates to the methods for their preparation, pharmaceutical compositions containing these compounds and uses of these compounds in the treatment of disorders/conditions/diseases involving, relating to or associated with enzymes having methyl transferase activities/functions and/or via unspecified/multi-targeted mechanisms.

Description

Description
Title of Invention: Compounds For Treatment Of Cancer and Epigenetics
Technical Field
The present invention generally relates to quinolines and 5,6,7,8-tetrahydroacridine derivatives, methods for their preparation, pharmaceutical compositions containing these compounds and uses of these compounds in the treatment of disorders/conditions/diseases involving, relating to or associated with enzymes having methyltransferase activities.
Background Art
The genomes of eukaryotic organisms are tightly packaged into chromatin, which forms the structural basis of nuclear processes associated with genetic activity. Nucleosome is the smallest structural unit of chromatin, in which 146 base pairs of DNA are wrapped around an octamer of core histones. The histones are subjected to several post-translational covalent modifications and this plays a critical role in controlling gene transcription within cells. Among the various histone modifications, methylation of lysine residue seems to play a particularly important role in control of gene transcription programs (Arrowsmith, C. H., et al. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discov. 2012, 11, 384-400). These modifications are catalyzed by a class of group-transfer enzymes known as the protein lysine methyltransferases (PKMT).
There are more than 100 protein methyltransferases that are encoded by the human genome and they are subdivided into five subfamilies based on their primary sequence. Misregulation of these proteins, through overexpression, deletion or chromosomal translocations has been implicated in many types of human cancers. PKMTs contain an evolutionarily conserved SET (Su(var), E(z) and Trithorax) domain that is involved in catalyzing the transfer of the methyl group from the cofactor 5- adenosyl-L-methionine (SAM) to lysine residues of histone and non-histone substrates, leading to lysine mono-, di-, and/or trimethylation. Histone lysine methylation has been increasingly recognized as a major epigenetic gene regulation mechanism in eukaryotic cells (Copeland, R. A., Molecular pathways: protein methyltransferases in cancer. Clin. Cancer Res. 2013, 19, 6344-6350).
SMYD3 is a histone methyltransferase and is overexpressed in several cancers including breast, gastric, pancreatic, colorectal, lung cancer and hepatocellular carcinoma. It tri-methylates histone H3 at lysine 4 (H3K4me3), a mark associated with gene activation. SMYD3 is part of the SMYD family (SET/MYND) of proteins which contains five members carrying a SET domain and a MYND type of zinc finger. The up-regulation of the SMYD3 promotes proliferation of HCC (hepatocellular carcinoma) via increasing H3 lysine 4 methylation, and a subsequent activation of downstream genes, including Nkx2.8 gene which is frequently up-regulated in human HCC (Hamamoto, R., et al. SMYD3 encodes a histone methyltransf erase involved in the proliferation of cancer cells. Nat. Cell Biol. 2004, 6, 731-740).
SMYD3 also catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of SMYD3 protein. It has been shown that the catalytic activity of SMYD3 is required for the anchorage-independent growth of cancer cells. Thus, SMYD3, via H4K5 methylation, provides another link between chromatin dynamics and neoplastic disease (Van Eller, et al. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics 2012, 7, 340-343).
SMYD3 modulates myostatin and c-Met transcription in primary skeletal muscle cells and C2C12 myogenic cells. It does this by targeting the myostatin and c-Met genes and participates in the recruitment of the bromodomain protein BRD4 to their regulatory regions through protein-protein interaction. By recruiting BRD4, SMYD3 favors chromatin engagement of the pause-release factor p- TEFb (positive transcription elongation factor) and elongation of Ser2-phosphorylated RNA polymerase II (PolIISer2P). SMYD3 is also known to methylate other substrates such as RBI protein (CA2613322 Al) and VEGFR1 (US8354223 B2).
It has been shown in mouse models of K-Ras-driven cancer, that SMYD3 acts in the cytoplasm of cancer cells, methylating a lysine residue (K260) on MAP3K2, a kinase enzyme that is associated with the activation of MEK-ERK mitogen-activated protein-kinase pathway (Mazur, P., et.al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature 2014, 510, 283- 287).
Early 2015, a SMYD3 inhibitor BCI-121 was reported by Peserico, A., et. al. (A SMYD3 small-molecule inhibitor impairing cancer cell growth. /. Cell Physiol. 2015, 230, 2447-2460). There was no reported biochemical assay data in the publication, and this inhibitor was found to be inactive against SMYD3 using the biochemical assay disclosed herein (Examples Section: Comparative Example 1).
Another prior art known includes tetrahydroacridine derivatives (WO 2011/086178) found to act against against a different target ubiquitin specific protease 7. The compounds disclosed in WO2011/086178 were found to act against a different target ubiquitin specific protease 7, but were not found to be active against SMYD3.
The following compounds are also known from Scifinder: ethyl 4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate and ethyl 4-(9-chloro-2,3-dihydro-lH- cyclopenta[b]quinoline-6-carbonyl)piperazine-l-carboxylate. Ethyl 4-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate was found to be moderately active against SMYD3, but suffered from poor metabolic stability due to high metabolic clearance in the human/mouse liver microsomes stability test. In addition, ethyl 4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate was found to have poor target engagement compared to a more advanced compound disclosed herein.
Recently, another small molecule inhibitor of SMYD3 was reported (Mitchell, L. H., et. al. Novel Oxindole Sulfonamides and Sulfamides: EPZ031686, the First Orally Bioavailable Small Molecule SMYD3 Inhibitor. ACS Med. Chem. Lett. 2015, ASAP). Although the reported molecule was active against SMYD3, no anti-proliferative cellular activity was disclosed. Moreover, the structure of the inhibitor is not related to the compounds in this application.
There is therefore an urgent need to provide novel compounds that overcome, or at least ameliorates, one or more of the disadvantages of the effects of protein lysine methyltransferases such as SMYD3 described above. There is also a need to provide a pharmaceutical composition comprising the compound, methods for treating diseases using the compound and a method for synthesizing the compound.
Summary
In a first aspect, there is rovided a compound having the following Formula (I);
Figure imgf000004_0001
(I)
wherein
Z1 and Z2 are independently selected from O, S or NH;
X is a halogen;
R1 and R2 are independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl , optionally substituted aryl and optionally substituted heteroaryl;
and wherein R1 and R2 may optionally be taken together to form an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S;
and wherein R1 and R2 may optionally form an optionally substituted aryl or optionally substituted heteroaryl together with the ring atoms that they are bonded to;
R3, R4, R5, R6, R7 and R8 are independently absent, or selected from the group consisting of a bond, hydrogen, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
wherein any two of R3, R4, R5, R6, R7 or R8 may be taken together to form an optionally substituted cycloalkyl or an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S ; Y is selected from R9, OR9 or NHR9, wherein R9 is an optionally substituted C3 to do alkyl, optionally substituted C3 to do alkenyl, optionally substituted C3 to do alkynyl, optionally substituted d to d cycloalkyl, optionally substituted d to do haloalkyl, a substituted 5-membered heteroaryl comprising two to three heteroatoms selected from N, O or S or a d to d alkyl substituted with an optionally substituted 5-membered heterocycloalkyl comprising one to two heteroatoms selected from N, O or S;
or a pharmaceutically acceptable form or prodrug thereof.
In an embodiment, there is provided the compound as defined above, having the following Formula (III):
Figure imgf000005_0001
(III)
wherein R1 and Rl la are independently selected from the group consisting of H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
Rnb may be absent, H or optionally substituted alkyl;
A2 is selected from CH, N, O or S; and
p is an integer selected from 0, 1 or 2.
In another embodiment, there is provided the compound as defined above, having the following Formula (IV):
Figure imgf000005_0002
(IV)
wherein
A3 and A4 are independently selected from CH or N;
A5 and A6 are independently selected from CH, N, O or S; R12a, R13a, R14 and R15 are independently selected from the group consisting of hydrogen, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
R12b and R13b are independently absent, H or an optionally substituted alkyl; and
q and r are independently integers selected from 0, 1 or 2.
Advantageously, the compound as defined above is an inhibitor of protein lysine methyltransferases (PKMT) such as SMYD3. SMYD3 is an attractive target for drug discovery due to its role in epigenetic regulation and crucial cell signalling pathways. Advantageously, a small molecule inhibitor of SMYD3 as defined above may be useful for the treatment of cancers with elevated SMYD3 expression such as hepatocellular carcinoma (HCC).
Advantageously, the compounds as defined above have a unique potency profile against the target protein. The compounds may be modified to have different potencies against different targets for a variety of indications or applications. More advantageously, the compound is a small molecule inhibitor. Small molecule inhibitors, unlike macromolecules such as polymers, proteins and DNA, may be less toxic and have fewer occurrences of adverse drug effects while maintaining a high level of activity.
Further advantageously, the tested compounds as defined above have a siginificantly higher potency against the target protein compared to conventionally known compounds. The compound as defined above has a significantly higher potency in inhibiting SMYD3.
In a second aspect, there is provided a pharmaceutical composition comprising a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, and a pharmaceutically acceptable excipient.
In a third aspect, there is provided a method of inhibiting SMYD3 in a cell comprising administering to a cell the compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above.
In a fourth aspect, there is provided a method of treating a SMYD3-related disorder comprising administering to a subject in need of treatment a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above.
In a fifth aspect, there is provided a use of a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above, in the manufacture of a medicament for treatment of a SMYD3-related disorder.
In a sixth aspect, there is provided a compound as defined above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as defined above, for use in the treatment of a SMYD3- related disorder.
Advantageously, the compounds as defined above have demonstrated inhibitory activities against the methyl transferase activity of SMYD3 enzyme and anti-proliferative activities against a variety of human tumor cell lines. The compound as defined above may demonstrate good drug-like properties, that is, in vitro metabolic stability, solubility and desirable lipophilicity. More advantageously, the compounds inhibit methyltransferase activity of SMYD3 in an MTase assay using MAP3K2 as a peptide substrate. Further advantageously, the compounds show antiproliferative activity. Further advantageously, the compounds inhibit SMYD3 mediated methylation of MAP3K2 and inhibit anchorage independent growth in human cancer cells.
In an eight aspect, there is provided a process for synthesizing the compound as defined above having the following Formula III), comprising the steps of:
Figure imgf000007_0001
(III)
(a) contacting an optionally substituted aminobenzoate ester with a compound having following Formula (Va) to form a cyclized product;
Figure imgf000007_0002
(Va)
wherein R16 is selected from the group consisting of H, methyl, COOMe and COOEt;
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form the compound of formula (III);
Figure imgf000007_0003
(VI)
wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
In a ninth aspect, there is provided a process for synthesizing the compound as defined above having the following Formula (III), wherein R1 is hydrogen; comprising the steps of:
Figure imgf000008_0001
(III)
(a) contacting an optionally substituted aminobenzoate ester with a compound having the following Formula (Vb) and phosphorus oxychloride to form a halogenated cyclized product;
Figure imgf000008_0002
(Vb),
(b) selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form an amide; and
Figure imgf000008_0003
(VI)
(c) selectively functionalizing at least one halogen of the halogenated cyclized product of step (a) with a group having the following formula (VII) under reaction conditions to form the compound of formula (III);
Figure imgf000008_0004
(VII)
wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
In a tenth aspect, there is provided a process for synthesizing the compound as defined above having the following formula (IV), comprising the steps of;
Figure imgf000009_0001
(IV)
(a) contacting an amino substituted terephthalic acid or an ester thereof; with an optionally substituted cyclic ketone having the following Formula (VIII) to form a cyclized product;
Figure imgf000009_0002
(VIII)
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) optionally selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid; and
(d) selectively functionalizing the carboxylic acid of the cyclized product of step (a) or (c) with a group having the following formula (VI) under reaction conditions to form the compound of formula (IV);
Figure imgf000009_0003
(VI)
wherein step (b), (c) and (d) may be performed simultaneously, sequentially or in any order.
Definitions
In this specification a number of terms are used which are well known to a skilled addressee. Nevertheless for the purposes of clarity a number of terms will be defined. The following words and terms used herein shall have the meaning indicated: In the definitions of a number of substituents below it is stated that "the group may be a terminal group or a bridging group". This is to signify that the use of the term is intended to encompass the situation where the group is a linker between two other portions of the molecule as well as where it is a terminal moiety. Using the term alkyl as an example, some publications would use the term "alkylene" for a bridging group and hence in these other publications there is a distinction between the terms "alkyl" (terminal group) and "alkylene" (bridging group). In the present application no such distinction is made and most groups may be either a bridging group or a terminal group.
"Acyl" means an R-C(=0)- group in which the R group may be an optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl or optionally substituted heteroaryl group as defined herein. Examples of acyl include acetyl, benzoyl and amino acid derived aminoacyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
"Acylamino" means an R-C(=0)-NH- group in which the R group may be an alkyl, cycloalkyl, heterocycloalkyl; aryl or heteroaryl group as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
"Aliphatic" means non-aromatic, open chain, straight or branched organic compounds.
"Alkenyl" as a group or part of a group denotes an aliphatic hydrocarbon group containing at least one carbon -carbon double bond and which may be straight or branched preferably having 2-12 carbon atoms, more preferably 2-10 carbon atoms, most preferably 2-6 carbon atoms, in the normal chain. The group may contain a plurality of double bonds in the normal chain and the orientation about each is independently E or Z. Exemplary alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl and nonenyl. The group may be a terminal group or a bridging group.
"Alkenyloxy" refers to an alkenyl-O- group in which alkenyl is as defined herein. Preferred alkenyloxy groups are C C6 alkenyloxy groups. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Alkyl" or "alkylene" as a group or part of a group refers to a straight or branched aliphatic hydrocarbon group, preferably a Q-Q2 alkyl, more preferably a Q-Qo alkyl, most preferably C C6 unless otherwise noted. Examples of suitable straight and branched Q-Ce alkyl substituents include methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, t-butyl, hexyl, and the like. The group may be a terminal group or a bridging group.
"Alkylamino" includes both mono-alkylamino and dialkylamino, unless specified. "Mono- alkylamino" means an Alkyl-NH- group, in which alkyl is as defined herein. "Dialkylamino" means a (alkyl)2N- group, in which each alkyl may be the same or different and are each as defined herein for alkyl. The alkyl group is preferably a C C6 alkyl group. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom. "Alkylaminocarbonyl" refers to a group of the formula (Alkyl)x(H)yNC(=0)- in which alkyl is as defined herein, x is 1 or 2, and the sum of X+Y =2. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
"Alkyloxy" refers to an alkyl-O- group in which alkyl is as defined herein. Preferably the alkyloxy is a C C6 alkyloxy. Examples include, but are not limited to, methoxy and ethoxy. The group may be a terminal group or a bridging group. The term alkyloxy may be used interchangeably with the term "alkoxy".
"Alkyloxy alkyl" refers to an alkyloxy-alkyl- group in which the alkyloxy and alkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Alkyloxyaryl" refers to an alkyloxy-aryl- group in which the alkyloxy and aryl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the aryl group.
"Alkyloxycarbonyl" refers to an alkyl-0-C(=0)- group in which alkyl is as defined herein. The alkyl group is preferably a Q-Ce alkyl group. Examples include, but are not limited to, methoxycarbonyl and ethoxycarbonyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the carbonyl carbon.
"Alkyloxycycloalkyl" refers to an alkyloxy-cycloalkyl- group in which the alkyloxy and cycloalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the cycloalkyl group.
"Alkyloxyheteroaryl" refers to an alkyloxy-heteroaryl- group in which the alkyloxy and heteroaryl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroaryl group.
"Alkyloxyheterocycloalkyl" refers to an alkyloxy-heterocycloalkyl- group in which the alkyloxy and heterocycloalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heterocycloalkyl group.
"Alkylsulfinyl" means an alkyl-S-(=0)- group in which alkyl is as defined herein. The alkyl group is preferably a C C6 alkyl group. Exemplary alkylsulfinyl groups include, but not limited to, methylsulfinyl and ethylsulfinyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
"Alkylsulfonyl" refers to an alkyl-S(=0)2- group in which alkyl is as defined above. The alkyl group is preferably a Q-Ce alkyl group. Examples include, but not limited to methylsulfonyl and ethylsulfonyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
"Alkynyl" as a group or part of a group means an aliphatic hydrocarbon group containing a carbon-carbon triple bond and which may be straight or branched preferably having from 2-12 carbon atoms, more preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms in the normal chain. Exemplary structures include, but are not limited to, ethynyl and propynyl. The group may be a terminal group or a bridging group.
"Alkynyloxy" refers to an alkynyl-O- group in which alkynyl is as defined herein. Preferred alkynyloxy groups are C C6 alkynyloxy groups. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Amino acid" as a group or part of a group means having at least one primary, secondary, tertiary or quaternary amino group, and at least one acid group, wherein the acid group may be a carboxylic, sulfonic, or phosphonic acid, or mixtures thereof. The amino groups may be "alpha", "beta", "gamma" ... to "omega" with respect to the acid group(s). The amino acid may be natural or synthetic, and may include their derivatives. The backbone of the "amino acid" may be substituted with one or more groups selected from halogen, hydroxy, guanido, heterocyclic groups. Thus the term "amino acids" also includes within its scope glycine, alanine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tryptophan, serine, threonine, cysteine, tyrosine, asparagine, glutamine, asparte, glutamine, lysine, arginine and histidine, taurine, betaine, N-methylalanine etc. (L) and (D) forms of amino acids are included in the scope of this disclosure. Additionally, the amino acids suitable for use in the present disclosure may be derivatized to include amino acids that are hydroxylated, phosphorylated, sulfonated, acylated, and glycosylated, to name a few.
"Amino acid residue" refers to amino acid structures that lack a hydrogen atom of the amino group (-NH-CHR-COOH), or the hydroxy moiety of the carboxy group (NH2-CHR-CO-), or both (- NH-CHR-CO-).
"Amino" refers to groups of the form -NRaRb wherein Ra and Rb are individually selected from the group including but not limited to hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, and optionally substituted aryl groups.
The terms "aminocarbonyl" group and "carbonylamino" group can be used interchangeably and are used to describe a -CO-NR2 group.
"Aminoalkyl" means an NH2-alkyl- group in which the alkyl group is as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Aminosulfonyl" means an NH2-S(=0)2- group. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
"Aryl" as a group or part of a group denotes (i) an optionally substituted monocyclic, or fused polycyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) preferably having from 6 to 12 atoms per ring. Examples of aryl groups include phenyl, naphthyl, and the like; (ii) an optionally substituted partially saturated bicyclic aromatic carbocyclic moiety in which a phenyl and a C5_7 cycloalkyl or C5_7 cycloalkenyl group are fused together to form a cyclic structure, such as tetrahydronaphthyl, indenyl or indanyl. The group may be a terminal group or a bridging group. Typically an aryl group is a C6-C18 aryl group. "Arylalkenyl" means an aryl-alkenyl- group in which the aryl and alkenyl are as defined herein. Exemplary arylalkenyl groups include phenylallyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
"Arylalkyl" means an aryl-alkyl- group in which the aryl and alkyl moieties are as defined herein. Preferred arylalkyl groups contain a C1 5 alkyl moiety. Exemplary arylalkyl groups include benzyl, phenethyl, 1-naphthalenemethyl and 2-naphthalenemethyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Arylalkyloxy" refers to an aryl-alkyl-O- group in which the alkyl and aryl are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Arylamino" includes both mono-arylamino and di-arylamino unless specified. Mono-arylamino means a group of formula arylNH-, in which aryl is as defined herein. Di-arylamino means a group of formula (aryl)2N- where each aryl may be the same or different and are each as defined herein for aryl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
"Arylheteroalkyl" means an aryl-heteroalkyl- group in which the aryl and heteroalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
"Aryloxy" refers to an aryl-O- group in which the aryl is as defined herein. Preferably the aryloxy is a Ce-Qg aryloxy, more preferably a C -Cw aryloxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Arylsulfonyl" means an aryl-S(=0)2- group in which the aryl group is as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
A "bond" is a linkage between atoms in a compound or molecule. The bond may be a single bond, a double bond, or a triple bond, as valency permits.
"Cycloaliphatic" means non-aromatic, cyclic organic compounds.
"Cycloalkenyl" means a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond and preferably having from 5-10 carbon atoms per ring. Exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl or cycloheptenyl. The cycloalkenyl group may be substituted by one or more substituent groups. A cycloalkenyl group typically is a C3-Q2 alkenyl group. The group may be a terminal group or a bridging group.
"Cycloalkyl" refers to a saturated monocyclic or fused or bridged or spiro polycyclic, carbocycle preferably containing from 3 to 9 carbon atoms per ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like, unless otherwise specified. It includes monocyclic systems such as cyclopropyl and cyclohexyl, bicyclic systems such as decalin, and polycyclic systems such as adamantane. A cycloalkyl group typically is a C3-Q2 alkyl group. The group may be a terminal group or a bridging group.
"Cycloalkylalkyl" means a cycloalkyl-alkyl- group in which the cycloalkyl and alkyl moieties are as defined herein. Exemplary monocycloalkylaikyl groups include cyclopropylmethyl, cyclopentylmethyl, cyclohexylmethyl and cycloheptylmethyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Cycloalkylalkenyl" means a cycloalkyl-alkenyl- group in which the cycloalkyl and alkenyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
"Cycloalkylheteroalkyl" means a cycloalkyl-heteroalkyl- group in which the cycloalkyl and heteroalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
"Cycloalkyloxy" refers to a cycloalkyl-O- group in which cycloalkyl is as defined herein. Preferably the cycloalkyloxy is a Ci-Cecycloalkyloxy. Examples include, but are not limited to, cyclopropanoxy and cyclobutanoxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Cycloalkenyloxy" refers to a cycloalkenyl-O- group in which the cycloalkenyl is as defined herein. Preferably the cycloalkenyloxy is a Q-Cecycloaikenyloxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Cycloamino" refers to a saturated monocyclic, bicyclic, or polycyclic ring containing at least one nitrogen atom in at least one ring. Each ring is preferably containing from 3 to 10 carbon atoms per ring, more preferably 4 to 7 carbon atoms per ring. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
"Haloalkyl" refers to an alkyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom selected from the group consisting of fluorine, chlorine, bromine and iodine. A haloalkyl group typically has the formula CnH(2n+i m)Xm wherein each X is independently selected from the group consisting of F, CI, Br and I . In groups of this type n is typically from 1 to 10, more preferably from 1 to 6, most preferably 1 to 3. m is typically 1 to 6, more preferably 1 to 3. Examples of haloalkyl include fluoromethyl, difluoromethyl and trifluoromethyl.
"Haloalkenyl" refers to an alkenyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom independently selected from the group consisting of F, CI, Br and I.
"Haloalkynyl" refers to an alkynyl group as defined herein in which one or more of the hydrogen atoms has been replaced with a halogen atom independently selected from the group consisting of F, CI, Br and I.
"Halogen" represents chlorine, fluorine, bromine or iodine. "Heteroalkyl" refers to a straight- or branched-chain alkyl group preferably having from 2 to 12 carbon atoms, more preferably 2 to 6 carbon atoms in the chain, one or more of which has been replaced by a heteroatom selected from S, O, P and N. Exemplary heteroalkyls include alkyl ethers, secondary and tertiary alkyl amines, amides, alkyl sulfides, and the like. Examples of heteroalkyl also include hydroxyQ-Ceaikyl, Q-CeaikyloxyQ-Ceaikyl, arninoQ-Ceaikyl, Q-CeaikylaminoQ-Ceaikyl, and di(C1-C6alkyl)aminoC1-C6alkyl. The group may be a terminal group or a bridging group.
"Heteroalkyloxy" refers to a heteroalkyl-O- group in which heteroalkyl is as defined herein. Preferably the heteroalkyloxy is a C C6 heteroalkyloxy. The group may be a terminal group or a bridging group.
"Heteroaryl" either alone or part of a group refers to groups containing an aromatic ring (preferably a 5 or 6 membered aromatic ring) having one or more heteroatoms as ring atoms in the aromatic ring with the remainder of the ring atoms being carbon atoms. Suitable heteroatoms include nitrogen, oxygen and sulphur. Examples of heteroaryl include thiophene, benzothiophene, benzofuran, benzimidazole, benzoxazole, benzothiazole, benzisothiazole, naphtho[2,3-b]thiophene, furan, isoindolizine, xantholene, phenoxatine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, tetrazole, indole, isoindole, lH-indazole, purine, quinoline, isoquinoline, phthalazine, naphthyridine, quinoxaline, cinnoline, carbazole, phenanthridine, acridine, phenazine, thiazole, isothiazole, phenothiazine, oxazole, isooxazole, furazane, phenoxazine, 2-, 3- or 4- pyridinyl,
2- , 3-, 4-, 5-, or 8- quinolinyl, 1-, 3-, 4-, or 5- isoquinolinyl 1-, 2-, or 3- indolyl, and 2-, or
3- thiophenyl. A heteroaryl group is typically a Q-Qg heteroaryl group. A heteroaryl group may comprise 3 to 8 ring atoms. A heteroaryl group may comprise 1 to 3 heteroatoms independently selected from the group consisting of N, O and S. The group may be a terminal group or a bridging group.
"Heteroarylalkyl" means a heteroaryl-alkyl group in which the heteroaryl and alkyl moieties are as defined herein. Preferred heteroarylalkyl groups contain a lower alkyl moiety. Exemplary heteroarylalkyl groups include pyridinylmethyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Heteroarylalkenyl" means a heteroaryl-alkenyl- group in which the heteroaryl and alkenyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
"Heteroarylheteroalkyl" means a heteroaryl-heteroalkyl- group in which the heteroaryl and heteroalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
"Heteroarylamino" refers to groups containing an aromatic ring (preferably 5 or 6 membered aromatic ring) having at least one nitrogen and at least another heteroatom as ring atoms in the aromatic ring, preferably from 1 to 3 heteroatoms in at least one ring. Suitable heteroatoms include nitrogen, oxygen and sulphur. Arylamino and aryl is as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom. "Heteroaryloxy" refers to a heteroaryl-O- group in which the heteroaryl is as defined herein. Preferably the heteroaryloxy is a Ci-Ci8heteroaryloxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Heterocyclic" refers to saturated, partially unsaturated or fully unsaturated monocyclic, bicyclic or polycyclic ring system containing at least one heteroatom selected from the group consisting of nitrogen, sulfur and oxygen as a ring atom. Examples of heterocyclic moieties include heterocycloalkyl, heterocycloalkenyl and heteroaryl.
"Heterocycloalkenyl" refers to a heterocycloalkyl as defined herein but containing at least one double bond. A heterocycloalkenyl group typically is a C2-Cn heterocycloalkenyl group. The group may be a terminal group or a bridging group.
"Heterocycloalkyl" refers to a saturated monocyclic, fused or bridged or spiro polycyclic ring containing at least one heteroatom selected from nitrogen, sulfur, oxygen, preferably from 1 to 3 heteroatoms in at least one ring. Each ring is preferably from 3 to 10 membered, more preferably 4 to 7 membered. Examples of suitable heterocycloalkyl substituents include pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiofuranyl, piperidinyl, piperazinyl, tetrahydropyranyl, morpholino, 1 ,3- diazapane, 1 ,4-diazapane, 1 ,4-oxazepane, and 1,4-oxathiapane. A heterocycloalkyl group typically is a C2-C12 heterocycloalkyl group. A heterocycloalkyl group may comprise 3 to 9 ring atoms. A heterocycloalkyl group may comprise 1 to 3 heteroatoms independently selected from the group consisting of N, O and S. The group may be a terminal group or a bridging group.
"Heterocycloalkylalkyl" refers to a heterocycloalkyl-alkyl- group in which the heterocycloalkyl and alkyl moieties are as defined herein. Exemplary heterocycloalkylalkyl groups include (2-tetrahydrofuranyl)methyl, (2-tetrahydrothiofuranyl)methyl. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkyl group.
"Heterocycloalkylalkenyl" refers to a heterocycloalkyl-alkenyl- group in which the heterocycloalkyl and alkenyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the alkenyl group.
"Heterocycloalkylheteroalkyl" means a heterocycloalkyl-heteroalkyl- group in which the heterocycloalkyl and heteroalkyl moieties are as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the heteroalkyl group.
"Heterocycloalkyloxy" refers to a heterocycloalkyl- O- group in which the heterocycloalkyl is as defined herein. Preferably the heterocycloalkyloxy is a Ci-Ceheterocycloalkyloxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom.
"Heterocycloalkenyloxy" refers to a heterocycloalkenyl-O- group in which heterocycloalkenyl is as defined herein. Preferably the heterocycloalkenyloxy is a C C6 heterocycloalkenyloxy. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the oxygen atom. "Heterocycloamino" refers to a saturated monocyclic, bicyclic, or polycyclic ring containing at least one nitrogen atom and at least another heteroatom selected from nitrogen, sulfur, oxygen, preferably from 1 to 3 heteroatoms in at least one ring. Each ring is preferably from 3 to 10 membered, more preferably 4 to 7 membered. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
"Hydroxyalkyl" refers to an alkyl group as defined herein in which one or more of the hydrogen atoms have been replaced with an OH group. A hydroxyalkyl group typically has the formula CnH(2n+i X)(OH)x In groups of this type, n is typically from 1 to 10, more preferably from 1 to 6, most preferably from 1 to 3. x is typically from 1 to 6, more preferably from 1 to 4.
"Lower alkyl" as a group means unless otherwise specified, an aliphatic hydrocarbon group which may be straight or branched having 1 to 6 carbon atoms in the chain, more preferably 1 to 4 carbon atoms such as methyl, ethyl, propyl (n-propyl or isopropyl) or butyl (n-butyl, isobutyl or t- butyl). The group may be a terminal group or a bridging group.
"Patient," as used herein, refers to an animal, preferably a mammal, and most preferably a human.
"Subject" refers to a human or an animal.
"Sulfinyl" means an R-S(=0)- group in which the R group may be OH, alkyl, cycloalkyl, heterocycloalkyl; aryl or heteroaryl group as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the sulfur atom.
"Sulfinylamino" means an R-S(=0)-NH- group in which the R group may be OH, alkyl, cycloalkyl, heterocycloalkyl; aryl or heteroaryl group as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
"Sulfonyl" means an R-S(=0)2- group in which the R group may be OH, alkyl, cycloalkyl, heterocycloalkyl; aryl or heteroaryl group as defined herein. The group may be a terminal group or a bridging group. If the group is a terminal group, it is bonded to the remainder of the molecule through the sulfur atom.
"Sulfonylamino" means an R-S(=0)2-NH- group. The group may be a terminal group or a bridging group. If the group is a terminal group it is bonded to the remainder of the molecule through the nitrogen atom.
It is understood that included in the family of compounds of Formula (I) are isomeric forms including diastereomers, enantiomers, tautomers, and geometrical isomers in "E" or "Z" configurational isomer or a mixture of E and Z isomers. It is also understood that some isomeric forms such as diastereomers, enantiomers, and geometrical isomers can be separated by physical and/or chemical methods and by those skilled in the art.
Some of the compounds of the disclosed embodiments may exist as single stereoisomers, racemates, and/or mixtures of enantiomers and /or diastereomers. All such single stereoisomers, racemates and mixtures thereof, are intended to be within the scope of the subject matter described and claimed.
Additionally, Formula (I) is intended to cover, where applicable, solvated as well as unsolvated forms of the compounds. Thus, each formula includes compounds having the indicated structure, including the hydrated as well as the non-hydrated forms.
Further, it is possible that compounds of the invention may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of the invention and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to indicate that a mixture of diastereomers is present.
The term "optionally substituted" as used herein means the group to which this term refers may be unsubstituted, or may be substituted with one or more groups independently selected from alkyl, alkenyl, alkynyl, thioalkyl, cycloalkyl, aminocycloalkyl, cycloalkylalkyl, cycloalkenyl, cycloalkylalkenyl, heterocycloalkyl, cycloalkylheteroalkyl, cycloalkyloxy, cycloalkylaminocarbonyl, cycloalkenyloxy, cycloamino, halogen, carboxyl, haloalkyl, haloalkenyl, haloalkynyl, alkynyloxy, heteroalkyl, heteroalkyloxy, hydroxyl, hydroxyalkyl, alkoxy, alkyloxyalkyloxyalkyl, cycloalkylalkyloxyalkyl, thioalkoxy, alkenyloxy, haloalkoxy, haloalkenyloxy, nitro, amino, aminocarbonyl, aminocarbonylalkyl, azidoalkyl, nitroalkyl, nitroalkenyl, nitroalkynyl, nitroheterocyclyl, alkylamino, alkylaminocarbonyl, dialkylamino, dialkylaminocarbonyl, alkenylamine, alkylcarbonylamino, aminoalkyl, alkynylamino, acyl, alkyloxy, alkyloxyalkyl, alkyloxyaryl, alkyloxycarbonyl, alkyloxycycloalkyl, alkyloxyheteroaryl, alkyloxyheterocycloalkyl, alkenoyl, alkynoyl, acylamino, diacylamino, acyloxy, alkylsulfonyloxy, heterocyclic, heterocycloalkenyl, heterocycloalkyl, heterocycloalkylalkyl, heterocycloalkylalkenyl, heterocycloalkylheteroalkyl, heterocycloalkyloxy, heterocycloalkenyloxy, heterocycloxy, heterocycloamino, haloheterocycloalkyl, alkylsulfinyl, alkylsulfonyl, alkylsulfenyl, alkylcarbonyloxy, alkylthio, acylthio, aminosulfonyl, phosphorus-containing groups such as phosphono and phosphinyl, sulfinyl, sulfinylamino, sulfonyl, sulfonylamino, alkylsulfamoyl, aryl, heteroaryl, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroalkyl, heteroarylamino, heteroaryloxy, arylalkenyl, arylalkyl, alkylaryl, alkylheteroaryl, aryloxy, arylsulfonyl, cyano, cyanate, isocyanate, -C(0)NH(alkyl), and - C(0)N(alkyl)2. The number of carbon and hetero atoms in the groups of the optional substituents is as defined for the groups below, e.g. an alkyl or alkylene moiety can be a C Cn alkyl.
Preferably, the halogen is chlorine, fluorine, bromine or iodine, the alkyl is an optionally substituted C Cn alkyl, the alkenyl is an optionally substituted C Cn alkenyl, the alkynyl is a Q-Q2 alkynyl, the thioalkyl is an optionally substituted C1-Q2 thioalkyl comprising 1 or 2 sulfur atoms, the alkyloxy is an optionally substituted Q-Ce alkyl-O- group, the cycloalkyl is an optionally substituted C3-C9 cycloalkyl, the aminocycloalkyl is an optionally substituted C3-C9 aminocycloalkyl, the cycloalkylalkyl is an optionally substituted C3 to C9 cycloalkylalkyl, the cycloalkenyl is an optionally substituted C3-C9 cycloalkenyl, the cycloalkylalkenyl is an optionally substituted C3 to C9 cycloalkylalkenyl, the heterocycloalkyl is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the cycloalkylheteroalkyl is an optionally substituted cycloalkylheteroalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the cycloalkyloxy is an optionally substituted cycloalkyloxy having a ring atom number of 3 to 8 and having 1 or 2 oxygen atoms, the cycloalkylaminocarbonyl is an optionally substituted cycloalkylaminocarbonyl having a ring number of 3 to 8 and having a-CO-NH2 group, the cycloalkenyloxy is an optionally substituted cycloalkenyloxy having a ring atom number of 3 to 8 and having 1 or 2 oxygen atoms, the cycloamino is an optionally substituted cycloamino having a ring atom number of 3 to 8 and having 1 or 2 nitrogen atoms, halo is selected from the group consisting of fluoro, chloro, bromo and iodo, haloalkyl is an optionally substituted Q-C12 haloalkyl having at least one halo group selected from the group consisting of fluoro, chloro, bromo and iodo, haloalkenyl is an optionally substituted C1-C12 haloalkenyl having at least one halo group selected from the group consisting of fluoro, chloro, bromo and iodo, haloalkynyl is an optionally substituted C1-C12 haloalkynyl having at least one halo group selected from the group consisting of fluoro, chloro, bromo and iodo, alkenyloxy is an optionally substituted C C6 alkenyloxy having at least one oxygen atom, alkynyloxy is an optionally substituted C C6 alkynyloxy having at least one oxygen atom, heteroalkyl is an optionally substituted C2-C12 alkyl having a least one heteroatom selected from the group consisting of N, O, P and S, heteroalkyloxy is an optionally substituted C2-C12 alkyl having at least one oxygen atom and at least one other heteroatom selected from the group consisting of N, O, P and S, hydroxyalkyl is a substituted alkyl having the formula CnH(2n+1_x)(OH)x where n is 1 to 10, the thioalkyloxy is an optionally substituted C C6 alkyl-O- group having at least one sulfur group, the haloalkyloxy is an optionally substituted C C6 alkyl-O- group having at least one other substituent selected from the group consisting of fluoro, chloro, bromo and iodo, haloalkenyloxy is an optionally substituted Q-Ce alkenyloxy having at least one oxygen atom and at least one other substituent selected from the group consisting of fluoro, chloro, bromo and iodo, the aminocarbonyl is a-CO-NH2 group, the aminocarbonylalkyl is an optionally substituted Ci to Q2 alkyl group having a -CO-NH2 group, the azidoalkyl is a C!-C12-alkyl-N3 group, the nitroalkyl is an optionally substituted Q-C12 alkyl having at least one nitro group, the nitroalkenyl is an optionally substituted C Cn alkenyl having at least one nitro group, the nitroalkynyl is an optionally substituted C Cn alkynyl having at least one nitro group, the nitroheterocyclyl is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and having at least one nitro group, the optionally substituted aryl is an optionally substituted Ce-Qs aryl, the heteroaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, alkylamino is an optionally substituted alkyl-NH- group having a C C6 alkyl group, dialkylamino is an optionally substituted (alkyl)2N- group having a Q-Ce alkyl group, alkylaminocarbonyl is an optionally substituted alkyl -NH-CO- group having a C C6 alkyl group, dialkylaminocarbonyl is an optionally substituted (alkyl)2N-CO- group having a C C6 alkyl group alkenylamine is an optionally substituted alkenyl-NH- group having a Q-Ce alkenyl group, alkylcarbonylamino is an optionally substaited C!-C12-alkyl-CO-NH2 group, alkynyl amino is an optionally substituted alkynyl-NH- group having a C C6 alkynyl group, alkyloxyalkyl is an optionally substituted alkyloxy group having a C C6 alkyl group, the alkyloxyalkyloxyalkyl is an optionally substituted Q to C6 alkyloxyalkyl substituted with a Q to C6 alkyloxyalkyl, cycloalkylalkyloxyalkyl is a Q to C alkyloxyalkyl substaituted with a C3 to C8 cycloalkyl group, alkyloxyaryl is an optionally substituted alkyloxy group having an optionally substituted Ce-Qs aryl, alkyloxycarbonyl is a an optionally substituted Q-C16 alkyloxy having a carbonyl group, alkyloxycyclocarbonyl is an optionally substituted optionally substituted C3 to C9 cycloalkylalkyl having a carbonyl group and an alkoxy group, the alkyloxyheteroaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and having a Q-Ce alkyloxy group, alkyloxyheterocycloalkyl is an optionally substituted an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S having a C C6 alkyloxy group, alkanoyl is an optionally substituted C Cn alkyl having a carbonyl group, alkenoyl is an optionally substituted C Cn alkenyl having a carbonyl group, alkynoyl is an optionally substituted C\-C\2 alkynyl having a carbonyl group, acylamino is an optionally substituted R-C(=0)-NH- group in which the R group may be a C\-C\2 alkyl, C3-Q2 cycloalkyl, C3-Q2 heterocycloalkyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, aryl having a ring atom number of 3 to 8 or heteroaryl group having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, diacylamino is an optionally substituted [R-C(=0)]2-NH group in which the R group may be a Q-Q2 alkyl, C3-C12 cycloalkyl, C3-C12 heterocycloalkyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S aryl having a ring atom number of 3 to 8 or heteroaryl group having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the acyloxy is a Q-Q2 acyloxy, the alkylsufonyloxy is an optionally substituted C C6 alkyl-O- group having at least one sulfonyl group, the alkylsulfamoyl is an optionally substituted C C6 alkyl-0 group having at last one sulfamoyl group, the heterocycloalkenyl is a heterocycloalkenyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heterocycloalkyl is a heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heterocycloalkylalkyl is an optionally substituted C3 to C9 cycloalkylalkyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heterocycloalkylalkenyl is an optionally substituted C3 to C9 cycloalkylalkenyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heterocycloalkylheteroalkyl is an optionally substituted C3 to C9 cycloalkylalkenyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heterocycloalkyloxy is an optionally substituted C to C9 cycloalkyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and an optionally substituted Q-Ce alkyl-O- group, the heterocycloalkenyloxy is an optionally substituted C to C9 cycloalknyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and an optionally substituted Q-Ce alkyl-O- group, the heterocycloxy is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a hydroxyl group, the heterocycloamino is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and an amino group, the haloheterocycloalkyl is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a halo group selected from the group consisting of fluoro, chloro, iodo and bromo, the alkylsulfinyl is an optionally substituted Q-C12 alkyl group having at least one sulfinyl group, the alkylsulfonyl is an optionally substituted C1-C12 alkyl group having at least one sulfonyl group, the alkylsulfenyl is an optionally substituted C1-C12 alkyl group having at least one sulfenyl group, the alkylcarbonyloxy is an optionally substituted Q-C12 alkyl group having at least one carbonyl group and at least one hydroxy group, the alkylthio is an optionally substituted Q-C12 alkyl group having at least one thiol group, the acylthio is R-C(=0)-S in which R group may be a Q-C12 alkyl, C3-Q2 cycloalkyl, C3-Q2 heterocycloalkyl having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, aryl having a ring atom number of 3 to 8 or heteroaryl group having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the heteroarylalkyl or alkylheteroaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a Q-C12 alkyl, the heteroarylalkenyl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a Q-C12 alkenyl, the heteroarylheteroalkyl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a C Cn alkyl having at least one heteroatom selected from the group consisting of N, O and S, the heteroarylamino is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and an amino group, the heteroaryloxy is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having at least one oxygen group, the arylalkenyl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a C Cn alkenyl, the arylalkyl or alkylaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S and a C1-Q2 alkyl, the aryloxy is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having at least one oxygen atom, or the arylsulfonyl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having at least one sulfur atom.
The term "pharmaceutically acceptable salts" refers to salts that retain the desired biological activity of the above -identified compounds, and include pharmaceutically acceptable acid addition salts and base addition salts. Suitable pharmaceutically acceptable acid addition salts of compounds of Formula (I) may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, sulfuric, and phosphoric acid. Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, heterocyclic carboxylic and sulfonic classes of organic acids, examples of which are formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, fumaric, maleic, alkyl sulfonic, arylsulfonic. Additional information on pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Co., Easton, PA 1995. In the case of agents that are solids, it is understood by those skilled in the art that the inventive compounds, agents and salts may exist in different crystalline or polymorphic forms, all of which are intended to be within the scope of the present disclosure and specified formulae.
"Prodrug" means a compound that undergoes conversion to a compound of formula (I) within a biological system, usually by metabolic means (e.g. by hydrolysis, reduction or oxidation). For example an ester prodrug of a compound of formula (I) containing a hydroxyl group may be converted by hydrolysis in vivo to the parent molecule. Suitable esters of compounds of formula (I) containing a hydroxyl group, are for example formates, acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-β- hydroxynaphthoates, gentisates, isethionates, di-/?-toluoyltartrates, methanesulfonates, ethanesulfonates , benzenesulfonates , p-toluenesulfonates, cyclohexylsulfamates, and quinates. As another example an ester prodrug of a compound of formula (I) containing a carboxy group may be convertible by hydrolysis in vivo to the parent molecule. (Examples of ester prodrugs are those described by FJ. Leinweber, Drug Metab. Res., 18:379, 1987). Similarly, an acyl prodrug of a compound of formula (I) containing an amino group may be converted by hydrolysis in vivo to the parent molecule (Many examples of prodrugs for these and other functional groups, including amines, are described in Prodrugs: Challenges and Rewards (Parts 1 and 2); Ed V. Stella, R. Borchardt, M. Hageman, R.Oliyai, H. Maag and J Tilley; Springer, 2007)
The term "therapeutically effective amount" or "effective amount" is an amount sufficient to effect beneficial or desired clinical results. An effective amount can be administered in one or more administrations. An effective amount is typically sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state.
The term "functional equivalent" is intended to include variants of the specific protein lysine methyl transferase species described herein. It will be understood that the protein lysine methyl transferases may have isoforms, such that while the primary, secondary, tertiary or quaternary structure of a given protein lysine methyl transferase isoform is different to the prototypical protein lysine methyl transferase, the molecule maintains biological activity as a protein lysine methyl transferase. Isoforms may arise from normal allelic variation within a population and include mutations such as amino acid substitution, deletion, addition, truncation, or duplication. Also included within the term "functional equivalent" are variants generated at the level of transcription. Enzymes have isoforms that arise from transcript variation. Other functional equivalents include protein lysine methyl transferases having altered post-translational modification such as glycosylation.
The term "reprogramming cells" is intended to include erasure and remodeling of epigenetic marks, such as DNA methylation, during mammalian development.
The word "substantially" does not exclude "completely" e.g. a composition which is "substantially free" from Y may be completely free from Y. Where necessary, the word "substantially" may be omitted from the definition of the invention.
Unless specified otherwise, the terms "comprising" and "comprise", and grammatical variants thereof, are intended to represent "open" or "inclusive" language such that they include recited elements but also permit inclusion of additional, unrecited elements.
As used herein, the term "about", in the context of concentrations of components of the formulations, typically means ±10% of the stated value, more typically ±7.5% of the stated value, more typically ± 5% of the stated value, more typically ± 4% of the stated value, more typically ± 3% of the stated value, more typically, ± 2% of the stated value, even more typically ± 1% of the stated value, and even more typically ± 0.5% of the stated value.
Throughout this disclosure, certain embodiments may be disclosed in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosed ranges. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
Certain embodiments may also be described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the disclosure. This includes the generic description of the embodiments with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.
Brief Description of Drawings
The accompanying drawings illustrate a disclosed embodiment and serves to explain the principles of the disclosed embodiment. It is to be understood, however, that the drawings are designed for purposes of illustration only, and not as a definition of the limits of the invention.
Fig.l
[Fig. 1] shows the effect of compounds on the methyltransferase activity of SMYD3 using MAP3K2 peptide as a substrate and refers to dose-response curves showing the effect of compounds on the methyltransferase activity of SMYD3 using MAP3K2 peptide as a substrate ; for compound A066 (Fig. 1A), for compound A088 (Fig. IB); for compound B019 (Fig. 1C); and for compound A074 (Fig. ID).
Fig.2
[Fig. 2] refers to dose -response curves showing that SMYD3 compounds inhibit the proliferation of HCC, Colorectal, Lung and Pancreatic carcinoma cell lines. Fig. 2A is a dose response curve showing inhibition of HepG2, Fig. 2B is a dose response curve showing inhibition of HCT116, Fig. 2C is the dose response curve showing inhibition of A549, Fig. 2D is the dose response curve showing inhibition of CFPAC-1 and Fig. 2E is a dose response curve showing inhibition of HPAF-II.
Fig.3
[Fig. 3] refers to an image of aWestern blot showing SMYD3 target engagement and inhibition of MAP3K2 methylation following treatment with 25.0 μΜ of compound B019 and X4, in HEK293 cells transiently transfected with Myc-SMYD3.
Fig.4
[Fig. 4] refers to colony images in 24-well plate and the corresponding dose response curves of 2 sets of experiments using HepG2 cells. Fig. 4A is a colony image and Fig. 4B is a dose response curve with compound A074. Fig. 4C is a colony image and Fig. 4D is a dose response curve with compound B019.
Detailed Description of Embodiments
The present disclosure provides a compound of the following Formula (I);
Figure imgf000024_0001
(I)
Z1 and Z2 may be selected from O, S or NH. Z1 may be O. Z2 may be O.
X may be a halogen. X may be chloro.
R1 and R2 may be independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
R1 and R2 may optionally be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
R1 and R2 may optionally form an optionally substituted aryl or optionally substituted heteroaryl together with the ring atoms that they are bond to.
R3, R4, R5, R6, R7 and R8 may be independently absent, or selected from the group consisting of a bond, H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
R3, R4, R5, R6, R7 and R8 may be independently hydrogen or methyl.
R3, R4, R5, R6, R7 or R8 may be taken together to form an optionally substituted cycloalkyl, or optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
Y may be selected from the group consisting of R9, OR9 or NHR9, wherein R9 is an optionally substituted C3 to C10 alkyl, optionally substituted C3 to C10 alkenyl, optionally substituted C3 to C10 alkynyl, optionally substituted C3 to C7 cycloalkyl, optionally substituted C2 to Ci0 haloalkyl, a substituted 5-membered heteroaryl comprising two or three heteroatoms selected from N, O or S or a Ci to C2 alkyl substituted with an optionally substituted 5-membered heterocycloalkyl comprising one to two heteroatoms selected from N, O or S.
The compound of Formula (I) may include a pharmaceutically acceptable form or prodrug thereof. The compound may have the following Formula (II):
Figure imgf000025_0001
(II).
The optionally substituted alkyl may be an optionally substituted Q-C12 alkyl. The optionally substituted alkyloxy may be an optionally substituted C Ci6 alkyloxy. The optionally substituted cycloalkyl may be an optionally substituted C3-C9 cycloalkyl. The optionally substituted heterocycloalkyl may be an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S , the optionally substituted aryl may be an optionally substituted Ce-Qg aryl, the optionally substituted heteroaryl may be an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the optionally substituted alkenyl may be an optionally substituted C2-Q2 alkenyl or the optionally substituted alkynyl may be an optionally substituted C2-Q2 alkynyl.
R9 may be a C3 to C10 alkyl, optionally substituted C3 to C6 alkenyl, optionally substituted C2 to C10 haloalkyl, or in each case C3 to C9 alkyl or C3 to C7 cycloalkyl, or substituted oxazolyl, isoxazolyl, 1,2-azole, pyrazolyl, triazolyl, or methylpyrrolidinonyl.
R9 may be selected from the group consisting of propyl, butyl, pentyl, -CH2CH(CH3)2, -CH2CH=CH,
2- fluoroethyl, 3-fluoropropyl, 5-cyclopropylisoxazol-3-yl, 5-isobutylisoxazol-3-yl, 5-methylisoxazol-
3- yl 5-methylpyrazol-3-yl, 1 -methyl- l,2,3-triazol-4-yl, l-cycloproyl-l,2,3-triazol-4-yl, 1-tert-butyl- l,2,3-triazol-4-yl, l-cyclopropyl-l,2-pyrazol-4-yl and (R)-pyrrolidin-2-onyl-5-methyl.
The compound may have the following formula (Ila):
Figure imgf000025_0002
A1 may be O or NH.
R10 may be a Q to C9 alkyl or a C to C7 cycloalkyl. R10 may be selected from the group consisting of methyl, isobutyl and cyclopropyl.
R1 and R2 may be independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted phenyl, optionally substituted pyrazolyl, optionally substituted thiazolyl, optionally substituted thiophenyl, optionally substituted benzo[d]imidazolyl, optionally substituted indolyl, optionally substituted isoindoyl, optionally substituted indazolyl, optionally substituted pyrrolyl, optionally substituted pyridinyl, optionally substituted benzyl, optionally substituted benzo[d]dioxolyl, optionally substituted benzotriazolyl, optionally substituted benzoxazolyl, optionally substituted benzofuranyl, optionally substituted pyrazolopyridinyl, optionally substituted pyrrolopyrimidinyl, optionally substituted pyrrolopyridinyl, optionally substituted naphthyridinyl, optionally substituted pyrimidinyl, optionally substituted benzothiazolyl, optionally substituted cyclopropyl, amino group optionally substituted with an optionally substituted phenyl and amino group optionally substituted with an optionally substituted pyridinyl.
The compound may have the following Formula (lib):
Figure imgf000026_0001
(lib)
R1 may be H or halogen. R2 may be selected from the group consisting of H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-hydroxyphenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 2-(trifluoromethyl)phenyl, 3-(trifluoromethyl)phenyl, 4- (trifluoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4-fluoromethylphenyl, 2- hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphenyl, 3-ethoxyethylphenyl, 4-ethoxyethylphenyl, 2-(azidomethyl)phenyl, 3-(azidomethyl)phenyl, 4-(azidomethyl)phenyl, 2,3-difluorophenyl, 2,4- difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3,5- difluoro-4-hydroxyphenyl, 3,5-difluoro-4-(aminocarbonyl)phenyl, 3,5-difluoro-4-aminomethylphenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-(cyanomethyl)phenyl, 3-(cyanomethyl)phenyl, 4- (cyanomethyl)phenyl, 2-nitrophenyl, 3-nitrophenyl, 4-nitrophenyl, 2-aminophenyl, 3-aminophenyl, 4- aminophenyl, 2-(aminomethyl)phenyl, 3-(aminomethyl)phenyl, 4-(aminomethyl)phenyl, 2- (dimethylamino)phenyl, 3-(dimethylamino)phenyl, 4-(dimethylamino)phenyl, 2-
(aminocarbonyl)phenyl, 3-(aminocarbonyl)phenyl, 4-(aminocarbonyl)phenyl, 2-
(methylaminocarbonyl)phenyl, 3-(methylaminocarbonyl)phenyl, 4-(methylaminocarbonyl)phenyl, 2- (ethylaminocarbonyl)phenyl, 3-(ethylaminocarbonyl)phenyl, 4-(ethylaminocarbonyl)phenyl, 4-(l- ethoxyethyl)phenyl, 4-(2-hydroxy-2-propyl)phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-methyl-3- pyridinyl, 4-methyl-3-pyridinyl, 5-methyl-3-pyridinyl, 6-methyl-3 -pyridinyl, 6-methoxycarbonyl-3- pyridinyl, thiophenyls such as 2-thiophenyl, 3-thiophenyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3- pyrrolidinyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, l-methyl-3-pyrrolyl, 3-(l,2,5-trimethyl)-pyrrolyl, 2- ethynylphenyl, 3-ethynylphenyl, 4-ethynylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-(l- hydroxyethyl)phenyl, 3-(l-hydroxyethyl)phenyl, 4-(l-hydroxyethyl)phenyl, 2-(2- hydroxyethyl)phenyl, 3 -(2-hydroxyethyl)phenyl, 4-(2-hydroxyethyl)phenyl, 4-fluoro-3 -methylphenyl, 4-fluoro-2-methylphenyl, 3-fluoro-2-methylphenyl, 3-fluoro-4-methylphenyl, 3-fluoro-5- methylphenyl, 2-fluoro-5 -methylphenyl, 4-fluoro-3-methoxyphenyl, 4-fluoro-2-methoxyphenyl, 3- fluoro-2-methoxyphenyl, 3-fluoro-4-methoxyphenyl, 3-fluoro-5-methoxyphenyl, 2-fluoro-5- methoxyphenyl, 4-fluoro-3-hydroxyphenyl, 4-fluoro-2-hydroxyphenyl, 4-hydroxy-3-fluorophenyl, 4- hydroxy-2 -fluorophenyl, 4-fluoro-3-hydroxymethylphenyl, 4-fluoro-2-hydroxymethylphenyl, 3- fluoro-2-hydroxymethylphenyl, 3-fluoro-4-hydroxymethylphenyl, 3-fluoro-5-hydroxymethylphenyl, 2-fluoro-5-hydroxymethylphenyl, 3-fluoro-4-(2-hydroxy-2-propyl)phenyl, 3-(aminocarbonyl)-4- fluorophenyl, 2-(aminocarbonyl)-4-fluorophenyl, 2-(aminocarbonyl)-3-fluorophenyl, 4- (aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-2- fluorophenyl, 4-fluoro-3-(methylaminocarbonyl)phenyl, 3-fluoro-4-(methylaminocarbonyl)phenyl, 4- fluoro-2-(methylaminocarbonyl)phenyl, 3-fluoro-2-(methylaminocarbonyl)phenyl, 4-
(cyclopropylaminocarbonyl)phenyl, 2-(cyclopropylaminocarbonyl)phenyl, 3 -
(cyclopropylaminocarbonyl)phenyl, 4-fluoro-3-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-4- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-2-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-2- (cyclopropylaminocarbonyl)phenyl, (3-fluoro-4-(dimethylaminocarbonyl)phenyl, 3-fluoro-5- (dimethylaminocarbonyl)phenyl, 2-fluoro-5-(dimethylaminocarbonyl)phenyl, 4-fluoro-3-
(dimethylaminocarbonyl)phenyl, 4-fluoro-2-(dimethylaminocarbonyl)phenyl, 3 -fluoro-2-
(dimethylaminocarbonyl)phenyl, 3-methyl-4-(methylaminocarbonyl)phenyl, 3-amino-4-fluorophenyl, 2-amino-4-fluorophenyl, 3-aminomethyl-4-fluorophenyl, 2-aminomethyl-4-fluorophenyl, 3- hydroxymethyl-4-methylphenyl, 2-hydroxymethyl-4-methyl -phenyl, 2-hydroxymethyl-3-methyl- phenyl, 4-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-2- methylphenyl, 2-morpholinophenyl, 3-morpholinophenyl, 4-morpholinophenyl, 2-(pyrrolidin-l- yl)phenyl, 3-(pyrrolidin- 1 -yl)phenyl, 4-(pyrrolidin- 1 -yl)phenyl, 4-( 1 -amino- 1 -cyclopropyl)phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2- methylthiazolyl, 4-methylthiazolyl, 4-(dimethylamido)phenyl, 2-(dimethylamido)phenyl, 3- (dimethylamido)phenyl, 2-benzylamin, 3-benzylamin, 4-benzylamin, 2-methylaminophenyl, 3- methylaminophenyl, 4-methylaminophenyl, 6-(l-methyl)indazolyl, 6-(2-methyl)indazolyl, 5-(l- methyl)indazolyl, 4-(l-methyl)indazolyl, 3-(l-methyl)indazolyl, 5-(2-methyl)indazolyl, 5-(3- methyl)indazolyl, 5-(l-methyl)pyrazolyl, 4-(l-methyl)-pyrazolyl, 3-(l-methyl) pyrazolyl, 4-(l- isopropyl)-pyrazolyl, 4-(l-difluoromethyl)-pyrazolyl, 4-(5-trifluoromethyl)-pyrazolyl, 4-(l-(2,2,2)- trifluoroethyl)pyrazolyl, 4-(l-cyclopentyl)pyrazolyl, 2-(l-methyl) pyrazolyl-phenyl, 3-(l-methyl) pyrazolyl-phenyl, 4-(imidazol-l-yl)phenyl, 1-imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-(4- methylpiperazino)phenyl, 3-(4-methylpiperazino)phenyl, 2-(4-methylpiperazino)phenyl, 3-[l,2,4]- triazol-4-ylphenyl, 2-[l,2,4]-triazol-4-yl phenyl, 4-[l,2,4]-triazol-4-ylphenyl, 3-(aminomethyl)-4- methoxyphenyl, 3-(aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-4-methoxyphenyl, 2- (aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-6-methoxyphenyl, 4-(aminomethyl)-3- methoxyphenyl, 2-(aminomethyl)-3-methoxyphenyl, 4-(dimethylaminomethyl)phenyl, 3- (dimethylaminomethyl)phenyl, 2-(dimethylaminomethyl)phenyl, 4-fluoro-3 -
(dimethylaminomethyl)phenyl, 4-fluoro-2-(dimethylaminomethyl)phenyl, 4-methoxy-3 -methylphenyl, 2-methoxy-4-methylphenyl, 3-methoxy-5-methylphenyl, 3-methoxy-4-methylphenyl, 2-methoxy-5- methylphenyl, 2-methoxy-6-methylphenyl, 2-methoxy-3 -methylphenyl, 4-methoxy-3- hydroxymethylphenyl, 3-methoxy-4-hydroxymethylphenyl, 2-methoxy-4-hydroxymethylphenyl, 3- methoxy-5-hydroxymethylphenyl, 2-methoxy-5-hydroxymethylphenyl, 2-methoxy-6- hydroxymethylphenyl, 2-methoxy-3-hydroxymethylphenyl, 4-hydroxy-3-hydroxymethylphenyl, 4- hydroxy-3 -methylphenyl, 3-ethoxy-4-hydroxyphenyl, 3-hydroxy-4-methylphenyl, 2-hydroxy-4- methylphenyl, 3-cyano-4-methylphenyl, 4-cyano-3 -methylphenyl, 2-cyano-4-methylphenyl, 3-cyano- 5-methylphenyl, 2-cyano-5-methylphenyl, 2-cyano-6-methylphenyl, 2-cyano-3-methylphenyl, 4- (aminosulfonyl)phenyl, 3-(aminosulfonyl)phenyl, 2-(aminosulfonyl)phenyl, 3-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 3-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-6-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-3-methoxyphenyl, 4-(N,N- dimethylaminomethyl)-3-methoxyphenyl, 3-(mo holinomethyl)phenyl, 2-(mo holinomethyl)phenyl, 4-(morpholinomethyl)phenyl, 3-cyano-4-methoxyphenyl, 2-cyano-4-methoxyphenyl, 3-cyano-5- methoxyphenyl, 2-cyano-5-methoxyphenyl, 2-cyano-6-methoxyphenyl, 2-cyano-3-methoxyphenyl, 4- cyano-3-methoxyphenyl, 4-aminomethyl-3-methylphenyl, 2- aminomethyl-4-methylphenyl, 3- aminomethyl-5-methylphenyl, 3-aminomethyl-4-methylphenyl, 2-aminomethyl-5-methylphenyl, 2- aminomethyl-6-methylphenyl, 2-aminomethyl-3-methylphenyl, (l-methyl)cyclopropyl, (2- methyl)cyclopropyl, 1-fluorocyclopropyl, 4-(2-methyl)pyridinyl, 3-(4-methyl)-pyridinyl, 2-(4- methyl)-pyridinyl, 2-(5-methyl)-pyridinyl, 2-(6-methyl)-pyridinyl, 2-(3-methyl)-pyridinyl, 2-(3- acetamido)-pyridinyl, 2-(4-acetamido)-pyridinyl, 2-(5-acetamido)-pyridinyl, 2-(6-acetamido)- pyridinyl, 3-(2-acetamido)-pyridinyl, 3-(4-acetamido)-pyridinyl, 3-(5-acetamido)-pyridinyl, 3-(6- acetamido)-pyridinyl, 4-(2-acetamido)-pyridinyl, 4-(3-acetamido)-pyridinyl, 4-(N- methylsulfamoyl)phenyl, 3-(N-methylsulfamoyl)phenyl, 2-(N-methylsulfamoyl)phenyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 3- (N-methylsulfamoyl)pyrrolyl, 3-(N,N-dimethylsulfamoyl)pyrrolyl, 4-(N-methylamido)phenyl, 3-(N- methylamido)phenyl, 2-(N-methylamido)phenyl, 4-(N-methylaminomethyl)phenyl, 3-(N- methylaminomethyl)phenyl, 2-(N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)-4- methoxyphenyl, 3-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-4- methoxyphenyl, 2-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-6- methoxyphenyl, 4-(N-methylaminomethyl)-3-methoxyphenyl, 2-(N-methylaminomethyl)-3- methoxyphenyl, 4-(acetylamino)phenyl, 3-(acetylamino)phenyl, 2-(acetylamino)phenyl, and ethynyl,
2- (5-N,N-dimethylaminomethyl)thiophenyl, 5-(2-methyl)indazolyl, 5-(3-methyl)indazolyl, 5-(7- methyl)indazolyl, 5-lH-indazolyl, 6-lH-indazolyl, 3-(l-methyl)pyrrolyl, 3-(2- methoxycarbonyl)pyrrolyl, 4-(2-methoxy)pyridinyl, 4-(lH-pyrrolo[2,3-b]pyridinyl), 5-(lH- pyrrolo[2,3-b]pyridinyl), 2-methyl-5-(lH-pyrrolo[2,3-b]pyridinyl), 4-(pyrazol-l-yl)phenyl, 4-(lH- pyrazol-5-yl)phenyl, 4-(lH-pyrazol-4yl)phenyl, 4-(lH-pyrazol-3-yl)phenyl, 4-carboxy-3- methylphenyl, 3-lH-pyrazolyl, 4-lH-pyrazolyl, 5-1H pyrazolyl, 4-lH-benzimidazolyl, 5-1H- benzimidazolyl, l-methyl-5— benzimidazolyl, 2-methyl-5-lH-benzimidazolyl, l-methyl-6- benzimidazolyl, 2-hydroxy-5-lH-benzimidazolyl, 5-(2-methyl)-benzoxazolyl, 5-(l-methyl)indolyl, 5- (3-methyl)indolyl, 4-lH-indazolyl, 3-(hydroxymethyl)phenyl, 3-hydroxyphenyl, l,3-benzodioxol-5-yl and l,2,3-benzotriazol-6-yl, 3-methyl-5-(lH-pyrazolo[3,4-b]pyridinyl, l-methyl-5-(lH-pyrrazolo[3,4- b]pyridinyl, 2-amino-5-pyrimidinyl, l,5-naphthyl-3-yl, l,5-naphthyridin-3-yl, 5-benzofuranyl, 6-(2- methyl)-benzothiazolyl, 5-(2-methyl)-benzothiazolyl, 5-benzoxazolyl, 6-benzoxazolyl, 6-(2-methyl)- benzoxazolyl, 5-(2-methyl)-benzoxazolyl, 4-((2-methoxyethoxy)methyl)phenyl, 4- (cyclopropylmethoxy)methyl)phenyl, 3-(2-(aminomethyl)-l,5-dimethyl)-pyrrolyl, 5-oxoisoindolinyl,
3- fluoro-4-pyrrolidin- 1 -yl-phenyl, 4-( 1 -aminocarbonylmethyl)-pyrazolyl, 4-(l -oxetan-3-yl)-pyrazolyl,
4- ( 1 -amino-2-methyl-2-propyl)phenyl, 4- 1 -(pyrrolidin- 1 -yl)ethyl)phenyl, 4-( 1 - dimethylamino)ethyl)phenyl, 4-(2-hydroxypropan-2-yl)phenyl, 4-(2-methyl, 1-methylamino-propan- 2-yl)phenyl, 4-(2-methyl, l-dimethylamino-propan-2-yl)phenyl, 4-(l-amino-2-hydroxypropan-2- yl)phenyl and 3-dimethylaminoethyl-4-methoxyphenyl.
The compound may have the following Formula (III):
Figure imgf000029_0001
(III)
R1 and Rlla may be independently selected from the group consisting of H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
Rnb may be absent, H or optionally substituted alkyl.
A2 may be selected from CH, N, O or S; and
p may be an integer selected from 0, 1 or 2.
When p is 0, the A2 linked group may represent Rlla or Rnb. When p is 0, the A2 linked group may represent Rl la.
The compound may have the following Formula (Ilia):
Figure imgf000029_0002
(Ilia).
R1 and Rlla may be independently selected from the group consisting of a bond, H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-hydroxyphenyl, 3- hydroxyphenyl, 4-hydroxyphenyl, 2-(trifluoromethyl)phenyl, 3-(trifluoromethyl)phenyl, 4- (trifluoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4-fluoromethylphenyl, 2- hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphenyl, 3-ethoxyethylphenyl, 4-ethoxyethylphenyl, 2-(azidomethyl)phenyl, 3-(azidomethyl)phenyl, 4-(azidomethyl)phenyl, 2,3-difluorophenyl, 2,4- difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3,5- difluoro-4-hydroxyphenyl, 3,5-difluoro-4-(aminocarbonyl)phenyl, 3,5-difluoro-4-aminomethylphenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-(cyanomethyl)phenyl, 3-(cyanomethyl)phenyl, 4- (cyanomethyl)phenyl, 2-nitrophenyl, 3-nitrophenyl, 4-nitrophenyl, 2-aminophenyl, 3-aminophenyl, 4- aminophenyl, 2-(aminomethyl)phenyl, 3-(aminomethyl)phenyl, 4-(aminomethyl)phenyl, 2- (dimethylamino)phenyl, 3-(dimethylamino)phenyl, 4-(dimethylamino)phenyl, 2-
(aminocarbonyl)phenyl, 3-(aminocarbonyl)phenyl, 4-(aminocarbonyl)phenyl, 2- (methylaminocarbonyl)phenyl, 3-(methylaminocarbonyl)phenyl, 4-(methylaminocarbonyl)phenyl, 2- (ethylaminocarbonyl)phenyl, 3-(ethylaminocarbonyl)phenyl, 4-(ethylaminocarbonyl)phenyl, 4-(l- ethoxyethyl)phenyl, 4-(2-hydroxy-2-propyl)phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-methyl-3- pyridinyl, 4-methyl-3-pyridinyl, 5-methyl-3-pyridinyl, 6-methyl-3-pyridinyl, 2-thiophenyl, 3- thiophenyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 1-methyl- 3-pyrrolyl, 3-(l,2,5-trimethyl)-pyrrolyl, 2-ethynylphenyl, 3-ethynylphenyl, 4-ethynylphenyl, 2- ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-(l -hydroxy ethyl)phenyl, 3-(l-hydroxyethyl)phenyl, 4- (1 -hydroxy ethyl)phenyl, 2-(2-hydroxyethyl)phenyl, 3-(2-hydroxyethyl)phenyl, 4-(2- hydroxyethyl)phenyl, 4-fluoro-(3-methyl)phenyl, 4-fluoro-(2-methyl)phenyl, 3-fluoro-(2- methyl)phenyl, 3-fluoro-(4-methyl)phenyl, 3-fluoro-(5-methyl)phenyl, 2-fluoro-(5-methyl)phenyl, 4- fluoro-(3-methoxy)phenyl, 4-fluoro-(2-methoxy)phenyl, 3-fluoro-(2-methoxy)phenyl, 3-fluoro-(4- methoxy)phenyl, 3-fluoro-(5-methoxy)phenyl, 2-fluoro-(5-methoxy)phenyl, 4-fluoro-3- hydroxyphenyl, 4-fluoro-2-hydroxyphenyl, 4-hydroxy-3 -fluorophenyl, 4-hydroxy-2-fluorophenyl, 4- fluoro-3-hydroxymethylphenyl, 4-fluoro-2-hydroxymethylphenyl, 3-fluoro-2-hydroxymethylphenyl, 3-fluoro-4-hydroxymethylphenyl, 3-fluoro-5-hydroxymethylphenyl, 2-fluoro-5-hydroxymethylphenyl, 3-fluoro-4-(2-hydroxy-2-propyl)phenyl, 3-(aminocarbonyl)-4-fluorophenyl, 2-(aminocarbonyl)-4- fluorophenyl, 2-(aminocarbonyl)-3-fluorophenyl, 4-(aminocarbonyl)-3-fluorophenyl, 5- (aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-2 -fluorophenyl, 4-fluoro-3-
(methylaminocarbonyl)phenyl, 3 -fluoro-4-(methylaminocarbonyl)phenyl, 4-fluoro-2-
(methylaminocarbonyl)phenyl, 3-fluoro-2-(methylaminocarbonyl)phenyl, 4-
(cyclopropylaminocarbonyl)phenyl, 2-(cyclopropylaminocarbonyl)phenyl, 3 -
(cyclopropylaminocarbonyl)phenyl, 4-fluoro-3-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-4- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-2-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-2- (cyclopropylaminocarbonyl)phenyl, (3-fluoro-4-(dimethylaminocarbonyl)phenyl, 3-fluoro-5- (dimethylaminocarbonyl)phenyl, 2-fluoro-5-(dimethylaminocarbonyl)phenyl, 4-fluoro-3-
(dimethylaminocarbonyl)phenyl, 4-fluoro-2-(dimethylaminocarbonyl)phenyl, 3 -fluoro-2-
(dimethylaminocarbonyl)phenyl, 3-methyl-4-(methylaminocarbonyl)phenyl, 3-amino-4-fluorophenyl,
2- amino-4-fluorophenyl, 3-aminomethyl-4-fluorophenyl, 2-aminomethyl-4-fluorophenyl, 3- hydroxymethyl-4-methylphenyl, 2-hydroxymethyl-4-methyl -phenyl, 2-hydroxymethyl-3-methyl- phenyl, 4-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-2- methylphenyl, 2-mo holinophenyl, 3-mo holinophenyl, 4-morpholinophenyl, 2-(l- pyrrolidinyl)phenyl, 3-(l-pyrrolidinyl)phenyl, 4-(l-pyrrolidinyl)phenyl, 4-(l-amino-l- cyclopropyl)phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-thiazolyl, 4-thiazolyl, 5- thiazolyl, 2-methylthiazolyl, 4-methylthiazolyl, 4-(dimethylamido)phenyl, 2-(dimethylamido)phenyl,
3- (dimethylamido)phenyl, 2-benzylamin, 3-benzylamin, 4-benzylamin, 2-methylaminophenyl, 3- methylaminophenyl, 4-methylaminophenyl, 6-(l-methyl)indazolyl, 6-(2-methyl)indazolyl, 5-(l- methyl)indazolyl, 5-(2-methyl)indazolyl, 4-(l-methyl)indazolyl, 3-(l-methyl)indazolyl, 5-(3- methyl)indazolyl, 5-(l-methyl)pyrazolyl, 4-(l-methyl)-pyrazolyl, 3-(l-methyl)pyrazolyl, 4-(l- isopropyl)-pyrazolyl, 4-(l-difluoromethyl)-pyrazolyl, 4-(5-trifluoromethyl)-pyrazolyl, 4-(l-(2,2,2)- trifluoroethyl)pyrazolyl, 4-(l-cyclopentyl)pyrazolyl, 2-(l-methyl)pyrazolylphenyl, 3-(l- methyl)pyrazolylphenyl, 1-imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-(imidazol-l-yl)phenyl, 4-(4- methylpiperazino)phenyl, 3-(4-methyl)piperazino)phenyl, 2-(4-methyl)piperazino)phenyl, 3-[l,2,4]- triazol-4-ylphenyl, 2- [ 1 ,2,4] -triazol-4-ylphenyl, 4- [ 1 ,2,4] -triazol-4-ylphenyl, 3-(aminomethyl) -4- methoxyphenyl, 3-(aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-4-methoxyphenyl, 2- (aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-6-methoxyphenyl, 4-(aminomethyl)-3- methoxyphenyl, 2-(aminomethyl)-3-methoxyphenyl, 4-(dimethylaminomethyl)phenyl, 3- (dimethylaminomethyl)phenyl, 2-(dimethylaminomethyl)phenyl, 4-fluoro-3 -
(dimethylaminomethyl)phenyl, 4-fluoro-2-(dimethylaminomethyl)phenyl, 4-methoxy-3 -methylphenyl, 2-methoxy-4-methylphenyl, 3-methoxy-5-methylphenyl, 3-methoxy-4-methylphenyl, 2-methoxy-5- methylphenyl, 2-methoxy-6-methylphenyl, 2-methoxy-3 -methylphenyl, 4-methoxy-3- hydroxymethylphenyl, 3-methoxy-4-hydroxymethylphenyl, 2-methoxy-4-hydroxymethylphenyl, 3- methoxy-5-hydroxymethylphenyl, 2-methoxy-5-hydroxymethylphenyl, 2-methoxy-6- hydroxymethylphenyl, 2-methoxy-3-hydroxymethylphenyl, 3-cyano-4-methylphenyl, 4-cyano-3- methylphenyl, 2-cyano-4-methylphenyl, 3 -cyano-5 -methylphenyl, 2-cyano-5 -methylphenyl, 2-cyano- 6-methylphenyl, 2-cyano-3-methylphenyl, 4-(aminosulfonyl)phenyl, 3-(aminosulfonyl)phenyl, 2- (aminosulfonyl)phenyl, 3-(N,N-dimethylaminomethyl)-4-methoxyphenyl, 3-(N,N- dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-4-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-6-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-3-methoxyphenyl, 4-(N,N-dimethylaminomethyl)-3-methoxyphenyl, 3- (morpholinomethyl)phenyl, 2-(mo holinomethyl)phenyl, 4-(mo holinomethyl)phenyl, 3-cyano-4- methoxyphenyl, 2-cyano-4-methoxyphenyl, 3-cyano-5-methoxyphenyl, 2-cyano-5 -methoxyphenyl, 2- cyano-6-methoxyphenyl, 2-cyano-3-methoxyphenyl, 4-cyano-3 -methoxyphenyl, 4-aminomethyl-3- methylphenyl, 2- aminomethyl-4-methylphenyl, 3 -aminomethyl-5 -methylphenyl, 3-aminomethyl-4- methylphenyl, 2-aminomethyl-5-methylphenyl, 2-aminomethyl-6-methylphenyl, 2-aminomethyl-3- methylphenyl, (l-methyl)cyclopropyl, (2-methyl)cyclopropyl, 1-fluorocyclopropyl, 4-(3- methyl)pyridinyl, 3-(4-methyl)pyridinyl, 2-(4-methyl)pyridinyl, 4-(2-methyl)pyridinyl, 2-(5- methyl)pyridinyl, 2-(6-methyl)pyridinyl, 2-(3-methyl)pyridinyl, 2-(3-acetamido)-pyridinyl, 2-(4- acetamido)-pyridinyl, 2-(5-acetamido)-pyridinyl, 2-(6-acetamido)-pyridinyl, 3-(2-acetamido)- pyridinyl, 3-(4-acetamido)-pyridinyl, 3-(5-acetamido)-pyridinyl, 3-(6-acetamido)-pyridinyl, 4-(2- acetamido)-pyridinyl, 4-(3-acetamido)-pyridinyl, 4-(N-methylsulfamoyl)phenyl, 3-(N- methylsulfamoyl)phenyl, 2-(N-methylsulfamoyl)phenyl, 4-(N-methylamido)phenyl, 3-(N- methylamido)phenyl, 2-(N-methylamido)phenyl, 4-(N,N-dimethylsulfamoyl)phenyl, 3-(N,N- dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 3-(N-methylsulfamoyl)pyrrolyl, 3- (N,N-dimethylsulfamoyl)pyrrolyl, 4-(N-methylaminomethyl)phenyl, 3-(N- methylaminomethyl)phenyl, 2-(N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)-4- methoxyphenyl, 3-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-4- methoxyphenyl, 2-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-6- methoxyphenyl, 4-(N-methylaminomethyl)-3-methoxyphenyl, 2-(N-methylaminomethyl)-3- methoxyphenyl, 4-(acetylamino)phenyl, 3-(acetylamino)phenyl, 2-(acetylamino)phenyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 2- (5-N,N-dimethylaminomethyl)thiophenyl, 5-(2-methyl)indazolyl, 5-(3-methyl)indazolyl, 5-(7- methyl)indazolyl, 5-lH-indazolyl, 6-lH-indazolyl, 3-(l-methyl)pyrrolyl, 3-(2- methoxycarbonyl)pyrrolyl, 4-(2-methoxy)pyridinyl, 4-(lH-pyrrolo[2,3-b]pyridinyl), 5-(lH- pyrrolo[2,3-b]pyridinyl), 2-methyl-5-(lH-pyrrolo[2,3-b]pyridinyl), 4-(pyrazol-l-yl)phenyl, 4-(lH- pyrazol-5-yl)phenyl, 4-(lH-pyrazol-4-yl)phenyl, 4-(lH-pyrazol-3-yl)phenyl, 4-carboxy-3- methylphenyl, 3-lH-pyrazolyl, 4-lH-pyrazolyl, 5-1H pyrazolyl, 4-lH-benzimidazolyl, 5-1H- benzimidazolyl, l-methyl-5-lH-benzimidazolyl, 2-methyl-5-lH-benzimidazolyl, l-methyl-6-lH- benzimidazolyl, 2-hydroxy-5-lH-benzimidazolyl, 5-(2-methyl)-benzoxazolyl, 5-(l-methyl)indolyl, 5- (3-methyl)indolyl, 4-lH-indazolyl, 3-(hydroxymethyl)phenyl, 3-hydroxyphenyl, l,3-benzodioxol-5-yl and l,2,3-benzotriazol-6-yl, 3-methyl-5-(lH-pyrazolo[3,4-b]pyridinyl, l-methyl-5-(lH-pyrrazolo[3,4- b]pyridinyl, 2-amino-5-pyrimidinyl, l,5-naphthyl-3-yl, 5-benzofuran, 6-(2-methyl)-benzothiazolyl, 5- (2-methyl)-benzothiazolyl, 5-benzoxazolyl, 6-benzoxazolyl, 6-(2-methyl)-benzoxazol, 4-((2- methoxyethoxy)methyl)phenyl, 4-(cyclopropylmethoxy)methyl)phenyl, 3-(2-(aminomethyl)-l,5- dimethyl)-pyrrolyl, 5 -oxoisoindolinyl, 3-fluoro-4-pyrrolidin- 1 -yl -phenyl, 4-( 1 -aminocarbonylmethyl)- pyrazolyl, 4-(l-oxetan-3-yl)-pyrazolyl, 4-(l-amino-2-methyl-2-propyl)phenyl, 4-l-(pyrrolidin-l- yl)ethyl)phenyl, and 4-(l-dimethylamino)ethyl)phenyl, 4-(2-hydroxypropan-2-yl)phenyl, 4-(2-methyl,
1- methylamino-propan-2-yl)phenyl, 4-(2-methyl, l-dimethylamino-propan-2-yl)phenyl, 4-(l-amino-
2- hydroxypropan-2-yl)phenyl and 3-dimethylaminoethyl-4-methoxyphenyl.
The compound may have the following Formula (Ilia'):
Figure imgf000032_0001
(Ilia').
R1 and Rlla may be independently selected from the group consisting of bond, H, cyano, methyl, ethyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4-methylphenyl, 2-(trifluoromethyl)phenyl, 3- (trifluoromethyl)phenyl, 4-(trifiuoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl,
2- fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4- methoxyphenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4- difluorophenyl, 3,5-difluorophenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 2- (cyanomethyl)phenyl, 3-(cyanomethyl)phenyl, 4-(cyanomethyl)phenyl, 2-nitrophenyl, 3-nitrophenyl, 4-nitrophenyl, 2-aminophenyl, 3-aminophenyl, 4-aminophenyl, 2-(aminomethyl)phenyl, 3- (aminomethyl)phenyl, 4-(aminomethyl)phenyl, 2-(dimethylamino)phenyl, 3-(dimethylamino)phenyl, 4-(dimethylamino)phenyl, 2-(aminocarbonyl)phenyl, 3-(aminocarbonyl)phenyl, 4- (aminocarbonyl)phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 5-methyl-3-pyridinyl, thiophenyls such as 2-thiophenyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 2-ethynylphenyl, 3-ethynylphenyl, 4- ethynylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 4-fluoro-(3-methyl)phenyl, 4-fluoro-(2- methyl)phenyl, 3-fluoro-(2-methyl)phenyl, 3-fluoro-(4-methyl)phenyl, 3-fluoro-(5-methyl)phenyl, 2- fluoro-(5-methyl)phenyl, 4-fluoro-(3-methoxy)phenyl, 4-fluoro-(2-methoxy)phenyl, 3-fluoro-(2- methoxy)phenyl, 3-fluoro-(4-methoxy)phenyl, 3-fluoro-(5-methoxy)phenyl, 2-fluoro-(5- methoxy)phenyl, 2-morpholinophenyl, 3-morpholinophenyl, 4-morpholinophenyl, 2-(l- pyrrolidinyl)phenyl, 3-(l-pyrrolidinyl)phenyl, 4-(l-pyrrolidinyl)phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-thiazolyl, 4-thiazolyl, 4-(dimethylamido)phenyl, 2-(dimethylamido)phenyl,
3- (dimethylamido)phenyl, 2-methylaminophenyl, 3-methylaminophenyl, 4-methylaminophenyl, 5-(l- methyl)indazolyl, 4-(l-methyl)indazolyl, 3-(l-methyl)indazolyl, 5-(l-methyl)pyrazolyl, 3-(l- methyl)pyrazolyl, 2-(l-methyl)pyrazolylphenyl, 3-(l-methyl)pyrazolylphenyl, 1 -imidazolyl, 2- imidazolyl, 3-imidazolyl, 4-(imidazol-l-yl)phenyl, 4-(N,N-dimethylsulfamoyl)phenyl, 3-(N,N- dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 4-(4-methylpiperazino)phenyl, 3-(4- methyl)piperazino)phenyl, 2-(4-methyl)piperazino)phenyl, 3 -[ 1 ,2,4] -triazol-4-ylphenyl, 2- [ 1 ,2,4] - triazol-4-ylphenyl, 4-[l,2,4]-triazol-4-ylphenyl, 3-(aminomethyl)-4-methoxyphenyl, 3- (aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-4-methoxyphenyl, 2-(aminomethyl)-5- methoxyphenyl, 2-(aminomethyl)-6-methoxyphenyl, 4-(aminomethyl)-3-methoxyphenyl, 2- (aminomethyl)-3-methoxyphenyl, 4-(dimethylaminomethyl)phenyl, 3-(dimethylaminomethyl)phenyl,
2- (dimethylaminomethyl)phenyl, 4-methoxy-3-methylphenyl, 2-methoxy-4-methylphenyl, 3- methoxy-5-methylphenyl, 3-methoxy-4-methylphenyl, 2-methoxy-5-methylphenyl, 2-methoxy-6- methylphenyl, 2-methoxy-3-methylphenyl, 4-methoxy-3-hydroxymethylphenyl, 3-methoxy-4- hydroxymethylphenyl, 2-methoxy-4-hydroxymethylphenyl, 3 -methoxy-5 -hydroxymethylphenyl, 2- methoxy-5-hydroxymethylphenyl, 2-methoxy-6-hydroxymethylphenyl, 2-methoxy-3- hydroxymethylphenyl, 3-cyano-4-methylphenyl, 4-cyano-3-methylphenyl, 2-cyano-4-methylphenyl,
3- cyano-5-methylphenyl, 2-cyano-5-methylphenyl, 2-cyano-6-methylphenyl, 2-cyano-3- methylphenyl, 4-(aminosulfonyl)phenyl, 3-(aminosulfonyl)phenyl, 2-(aminosulfonyl)phenyl, 3-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 3-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-6-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-3-methoxyphenyl, 4-(N,N- dimethylaminomethyl)-3-methoxyphenyl, 3-(mo holinomethyl)phenyl, 2-(morpholinomethyl)phenyl,
4- (morpholinomethyl)phenyl, 3-cyano-4-methoxyphenyl, 2-cyano-4-methoxyphenyl, 3-cyano-5- methoxyphenyl, 2-cyano-5-methoxyphenyl, 2-cyano-6-methoxyphenyl, 2-cyano-3-methoxyphenyl, 4- cyano-3-methoxyphenyl, 4-aminomethyl-3-methylphenyl, 2- aminomethyl-4-methylphenyl, 3- aminomethyl-5-methylphenyl, 3-aminomethyl-4-methylphenyl, 2-aminomethyl-5-methylphenyl, 2- aminomethyl-6-methylphenyl, 2-aminomethyl-3-methylphenyl, (l-methyl)cyclopropyl, (2- methyl)cyclopropyl, 4-(3-methyl)pyridinyl, 3-(4-methyl)pyridinyl, 2-(4-methyl)pyridinyl, 4-(2- methyl)pyridinyl, 2-(5-methyl)pyridinyl, 2-(6-methyl)pyridinyl, 2-(3-methyl)pyridinyl, 4-(N- methylsulfamoyl)phenyl, 3-(N-methylsulfamoyl)phenyl, 2-(N-methylsulfamoyl)phenyl, 4-(N- methylamido)phenyl, 3-(N-methylamido)phenyl, 2-(N-methylamido)phenyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 4- (N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)phenyl, 2-(N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)-4-methoxyphenyl, 3-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N- methylaminomethyl)-4-methoxyphenyl, 2-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N- methylaminomethyl)-6-methoxyphenyl, 4-(N-methylaminomethyl)-3-methoxyphenyl, 2-(N- methylaminomethyl)-3-methoxyphenyl, 4-(acetylamino)phenyl, 3-(acetylamino)phenyl, 2- (acetylamino)phenyl, 4-(N,N-dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N- dimethylsulfamoyl)phenyl, 2-(5-N,N-dimethylaminomethyl)thiophenyl, 5-(2-methyl)indazolyl, 5-1H- indazolyl, 6-lH-indazolyl, 3-(l-methyl)pyrrolyl, 4-(2-methoxy)pyridinyl, 5-(lH-pyrrolo[2,3- b]pyridinyl), 4-(pyrazol-l-yl)phenyl, 4-(lH-pyrazol-5-yl)phenyl, 4-(lH-pyrazol-4-yl)phenyl, 4-(lH- pyrazol-3-yl)phenyl, 4-carboxy-3-methylphenyl, 3-lH-pyrazolyl, 5-lH-benzimidazolyl, 5-(l- methyl)indolyl, 4-lH-indazolyl, 3-(hydroxymethyl)phenyl and 3-hydroxyphenyl and ethynyl.
In some embodiments, R1 and R2 may be taken together to form an optionally substituted 5-membered cycloalkyl, an optionally substituted 6-membered cycloalkyl, an optionally substituted 5-membered heterocycloalkyl or an optionally substituted 6-membered heterocycloalkyl. The compound may have the following Formula (IV):
Figure imgf000034_0001
(IV).
A3 and A4 may be independently selected from CH or N.
A5 and A6 may be independently selected from CH, N, O or S.
R12a, R13a, R14 and R15 may be independently selected from the group consisting of H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
R12b and R13b may be independently absent, H or an optionally substituted alkyl; and
q and r may be independently integers selected from 0, 1 or 2.
When q is 0, the A5 linked group may represent R12a or R12b. When q is 0, the A5 linked group may represent R12a.
When r is 0, the A6 linked group may represent R13a or R13b. When r is 0, the A6 linked group may represent R13a.
A3 and A4 may be both C.
The compound may have the following Formula (IV a):
Figure imgf000034_0002
(IVa)
wherein R9 may be an optionally substituted C3 to C10 alkyl, optionally substituted C3 to C10 alkenyl, optionally substituted C3 to C10 alkynyl or optionally substituted C3 to C7 cycloalkyl.
R12a, R13a, R14 and R15 may be independently selected from H, methyl, ethyl, propyl, butyl, halogen, cyano, COOMe, COOEt, phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-(3-methyl)pyridinyl, 2-(4- methyl)pyridinyl, 2-(5-methyl)pyridinyl, 2-(6-methyl)pyridinyl, 3-(2-methyl)pyridinyl, 3-(4- methyl)pyridinyl, 3-(5-methyl)pyridinyl, 3-(6-methyl)pyridinyl, 4-(2-methyl)pyridinyl, 4-(3- fluoro)pyridinyl, 2-(3-fluoro)pyridinyl, 2-(4-fluoro)pyridinyl, 2-(5-fluoro)pyridinyl, 2-(6- fluoro)pyridinyl, 3-(2-fluoro)pyridinyl, 3-(4-fluoro)pyridinyl, 3-(5-fluoro)pyridinyl, 3-(6- fluoro)pyridinyl, 4-(2-fluoro)pyridinyl, 4-(3-fluoro)pyridinyl, 2-(3-cyano)pyridinyl, 4-(2- cyano)pyridinyl, 2-(5-cyano)pyridinyl, 2-(6-cyano)pyridinyl, 3-(2-cyano)pyridinyl, 3-(4- cyano)pyridinyl, 3-(5-cyano)pyridinyl, 3-(6-cyano)pyridinyl, 4-(2-cyano)pyridinyl, 2-[3- (aminocarbonyl)]pyridinyl, 2-[4-(aminocarbonyl)]pyridinyl, 2-[5-(aminocarbonyl)]pyridinyl, 2-[6- (aminocarbonyl)]pyridinyl, 3-[2-(aminocarbonyl)]pyridinyl, 3-[4-(aminocarbonyl)]pyridinyl, 3-[5- (aminocarbonyl)]pyridinyl, 3-[6-(aminocarbonyl)]pyridinyl, 4-[2-(aminocarbonyl)]pyridinyl, 2-[3- (aminomethyl)]pyridinyl, 2-[4-(aminomethyl)]pyridinyl, 2-[5-(aminomethyl)]pyridinyl, 2-[6- (aminomethyl)]pyridinyl, 4-[2-(aminomethyl)]pyridinyl, 3-[4-(aminomethyl)]pyridinyl, 3-[5- (aminomethyl)]pyridinyl, 3-[6-(aminomethyl)]pyridinyl, 4-[2-(aminomethyl)]pyridinyl, 2-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-thiazolyl, 3-thiazolyl, pyrrolidine, 5-methyl-l,2,4-oxadiazol-3-yl, NH2, -CH=CH2, CH2NH2, CH2CH2NH2, CH2N(CH3)2, C(0)NH2, NHC(NH)NH2,CH2NHC(NH)NH2, N(CH3)2, CH2CH=CH2, ethynyl and 4-(3-methyl)pyrimidinyl, 2-(4-ethynyl)pyridinyl, 3-(4- ethynyl)pyridinyl, 2-(6-ethynyl)pyridinyl, 2-(5-ethynyl)pyridinyl, 3-(4-ethynyl)pyridinyl, 3-(2- ethynyl)pyridinyl, 3-(5-ethynyl)pyridinyl, 3-(6-ethynyl)pyridinyl, 2-(3-cyano)pyrimidinyl, 2-(5- cyano)pyrimidinyl, 2-(6-cyano)pyrimidinyl, 3-(2-cyano)pyrimidinyl, 2-(N-methyl)pyrazolyl, 3-(N- methyl)pyrazolyl, CH2-pyrrolidine, and CH2CH2-pyrrolidine.
R3, R4, R5, R6, R7 and R8 may be independently selected from the group consisting of a bond, H, methyl, (S)-methyl, (R)-methyl, ethyl, (S)-ethyl, (R)-ethyl, cyano, -CH2OH, (S)-CH2OH, (R)-CH2OH, COOCH3, (S)-COOCH3, (R)-COOCH3, CH2OC(0)CH3, (R)-CH2OC(0)CH3, (S)-CH2OC(0)CH3, CH2OC(0)CH2CH2OCH3, (R)-CH2OC(0)CH2CH2OCH3, (S)-CH2OC(0)CH2CH2OCH3, CH2CH2OH, (R)-CH2CH2OH, (S)-CH2CH2OH, CH2OC(0)CH2CH2CH3, (R)-CH2OC(0)CH2CH2CH3, (S)- CH2OC(0)CH2CH2CH3, CF3, (R)-CF3, (S)-CF3, CH2OCH3, (S)-CH2OCH3, (R)-CH2OCH3, CONHCH3, (S)-CONHCH3, (R)-CONHCH3> CH2CONHCH3, (S)-CH2CONHCH3, (R)- CH2CONHCH3j CH2COOCH3, (S)-CH2COOCH3, (R)-CH2COOCH3, CH2OC(0)CH(CH3)2, (S)- CH2OC(0)CH(CH3)2, (R)-CH2OC(0)CH(CH3)2, CONH2, (S)-CONH2, (R)-CONH2, CH2CON(CH3)2, (S)-CH2CON(CH3)2, (R)-CH2CON(CH3)2, and CH2C(0)NH(CH3).
R4 and R5 or R6 and R7 may be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
R4 and R5 or R6 and R7 may be taken together to form a cyclopropane.
R3 and R4, R3 and R5, R3 and R6, R3 and R7, R3 and R8, R4 and R6, R4 and R7, R4 and R8, R5 and R6, R5 and R 7', R' and R 8°, R 6u and R 8 or R 7 and R 8° may be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S. The alkylene bridge may be a 1-carbon, 2-carbon or 3-carbon alkylene bridging group wherein -CH- units may optionally be replaced with -NH-, O or S.
For the substituents in Formula (I), (II), (III) and (IV) it can be further stated:
7 and Z2 preferably represent O. X preferably represents chlorine, bromine or fluorine. R1 and R2 independently from another preferably represent of a bond, H, cyano, Q-C4 alkyl, in each case optionally cyano, fluorine, chlorine, bromine, amino, di(Q- C6 alkyl ) amino, nitro, carbamoyl, Q- C6 alkyl, Q-C6 haloalkyl, C3-C6 alkene, C3-C6 alkyne, Q-C4 alkoxy, Q-C4 alkyl-CN, Q-C4- alkylamino, C1-C4 alkyl-CO-NH2, C1-C4 alkyl-NH-C(NH)-NH2, C1-C4 alkyl-C3-C6 cycloalkyl, Q- C4 alkylamino, Q-C4 alkyl- di (Ci-C3-alkyl) amino, Q-C4 alkoxycarbonyl, pyrrolidinyl substituted phenyl, phenylamino, benzyl, pyridinyl, pyridinylamino, pyrimidinyl or pyrimidinylamino.
R1 and R2 preferably may optionally be taken together to form a optionally substituted C3-C5 alkylene bridge wherein one -CH2- unit may be replaced with -NH-, S or O and wherein the optional substituents of the alkylene bridge are selected from cyano, fluorine, chlorine, bromine, carbamoyl, Q-C6 alkyl, Q-Q haloalkyl, C3- C6 alkene, C3-C6 alkyne, Q-C4 alkoxy, C C4 alkyl-CN, C C4 alkylamino, Q-C4 alkyl-CO-NH2, Q-C4 alkyl-NH-C(NH)-NH2, Q-C4 alkyl-C3-C6 cycloalkyl, C C4 alkylamino, Q-C4 alkyl-di(Ci-C alkyl) amino, C1-C4 alkyl-CO-Ci-C4 alkyl, C1-C4 alkoxycarbonyl; in each case optionally cyano, fluorine, chlorine, bromine, carbamoyl, Q-Ce alkyl, Q- C haloalkyl, C3-C6 alkene, C3-C6 alkyne, C1-C4 alkoxy, C1-C4 alkyl-CN, C C4 alkylamino, C1-C4 alkyl-CO-NH2, C1-C4 alkyl-NH-C(NH)-NH2, C1-C4 alkyl-C3-C6 cycloalkyl, C1-C4 alkylamino, C1-C4 alkyl- di (C C3 alkyl) amino, Q- C4 alkoxycarbonyl phenyl, benzyl or pyrrolidinyl; or in each case optionally cyano, fluorine, chlorine, bromine, carbamoyl, Q- C6 alkyl, C C6 haloalkyl, C3-C6 alkene, C3-C6 alkyne, d-C4 alkoxy, C C4 alkyl-CN, Q-C4 alkylamino, Q-C4 alkyl-CO-NH2, d-C4 alkyl-NH-C(NH)-NH2, Q-C4 alkyl-C3-C6cycloalkyl, Q-C4 alkylamino, Q-C4 alkyl-d^Q-Q alkyl) amino, Q-C4 alkoxycarbonyl substituted four or six membered heteroaryl or Q-C4 alkyl-heteroaryl containing one to three heteroatoms selected from O, N, or S.
Most preferably R1 and R2 form an optionally substituted -CH2-CH2-CH2-CH2- bridge.
R3, R4, R5, R6, R7 and R8 are independently absent, or represent a bond, H, cyano, carbamoyl, -COO- C1-C4 alkyl, -CO-NH- C1-C4 alkyl, C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 alkyl-OH, C1-C4 alkyl-CO- NH2, C1-C4 alkyl-CO- C1-C4 alkyl, Q- C4 alkyl-CO- di (C C3 alkyl) amino, C1-C4 alkyl-CO- NH- Q- C3 alkyl, d-C4 alkyl-O-CO- CrC4 alkyl or Q- C4 alkyl-O-CO- C C3 alkyl-O- Q-Q-alkyl. R3, R4, R5, R6, R7 or R8 may also be taken together to form a Q-C4 alkylene bridge. Most preferably two R at the same carbon atom form a -CH2-CH2- bridge or two R at different carbon atoms form a -CH2-CH2- CH2- bridge. Y preferably represents R9, OR9, or NH-R9 wherein R9 represents C3-C6 alkyl, C3-C6 alkenyl, optionally substituted C3-C10 alkynyl or a in each case optionally C C6 alkyl or C3-C6 cycloalkyl substituted isoxazolyl, oxazolyl or pyrazolyl. Y most preferably is OR9, if R9 is C3-C6 alkyl, C3-C6 alkenyl, optionally substituted C3-C10 alkynyl. Y most preferably is R9, if R9 is a in each case optionally Q-Ce alkyl or C -Ce cycloalkyl substituted isoxazolyl, oxazolyl or pyrazolyl.
R9 is preferably C to C9 alkyl, optionally substituted C to Ce alkenyl or a C to C9 alkyl, or C to C7 cycloalkyl substituted oxazolyl, isoxazolyl or pyrazolyl.
R9 is more preferably selected from the group consisting of propyl, butyl, pentyl, -CH2CH(CH3)2, - CH2CH=CH, 5-cyclopropylisoxazol-3-yl, 5-isobutylisoxazol-3-yl, 5-methylisoxazol-3-yl and 5- methylpyrazol-3-yl. R9 most preferably is propyl.
Compounds wherein R1 is preferably methyl or H, most preferably H, may be specifically mentioned. The compound may be selected from the group consisting of: 
Figure imgf000037_0001

Figure imgf000038_0001

Figure imgf000039_0001

Figure imgf000040_0001
40
Figure imgf000041_0001
41
Figure imgf000042_0001
42
Figure imgf000043_0001
43
Figure imgf000044_0001
44
Figure imgf000045_0001

Figure imgf000046_0001

Figure imgf000047_0001

Figure imgf000048_0001

Figure imgf000049_0001

Figure imgf000050_0001
50
Figure imgf000051_0001
51
Figure imgf000052_0001
52
Figure imgf000053_0001
53
Figure imgf000054_0001
54
Figure imgf000055_0001
Figure imgf000055_0002
55
Figure imgf000056_0001
56
Figure imgf000057_0001
Figure imgf000058_0001
The compound is an enzyme inhibitor. In certain embodiments, the compounds may be a protein lysine methyltransferase (PKMT) inhibitor. It may be an inhibitor for SET domain -containing and non-SET domain-containing methyl transferases. In particular, the protein lysine methyltransferase may be SMYD3. SMYD3 may also methylate other substrates such as the retinoblastoma (RB I) protein or the vascular endothelial growth factor receptor 1 (VEGFR1) protein.
The compound inhibits methylation of a histone. The histone may be of the HI, H2A, H2B, H3 or H4 family. The histone may be of the H1F, H1H1, H2AF, H2A1, H2A2, H2BF, H2B 1, H2B2, H3A1, H3A2, H3A3, H41 or H44 subfamily. The histone may be H1F0, H1FNT, H1FOO, H1FX, HIST1H1A, HIST1H1B, HIST1H1C, HIST1H1D, HIST1H1E, HIST1H1T, H2AFB1, H2AFB2, H2AFB3, H2AFJ, H2AFV, H2AFX, H2AFY, H2AFY2, H2AFZ, HIST1H2AA, HIST1H2AB, HIST1H2AC, HIST1H2AD, HIST1H2AE, HIST1H2AG, HIST1H2AI, HIST1H2AJ, HIST1H2AK, HIST1H2AL, HIST1H2AM, HIST2H2AA3, HIST2H2AC, H2BFM, H2BFS, H2BFWT, HIST1H2BA, HIST 1H2BB, HIST 1H2BC, HIST1H2BD, HIST1H2BE, HIST1H2BF, HIST1H2BG, HIST1H2BH, HIST1H2BI, HIST1H2BJ, HIST1H2BK, HIST1H2BL, HIST1H2BM, HIST1H2BN, HIST1H2BO, HIST2H2BE, HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3L HIST1H3J, HIST2H3C, HIST3H3, HIST1H4A, HIST1H4B, HIST1H4C, HIST1H4D, HIST1H4E, HIST1H4F, HIST1H4G, HIST1H4H, HIST1H4I, HIST1H4J, HIST1H4K, HIST1H4L, HIST4H4.
The compound inhibits methylation of histone by inhibiting lysine methyltransferases. The compound may inhibit ASH1L, DOT1L, EHMT1, EHMT2, EZH1, EZH2, MLL, MLL2, MLL3, MLL4, MLL5, NSD1, NSD2, NSD3, PRDM2, PRDM9, SET, SETBP1, SETD1A, SETD1B, SETD2, SETD3, SETD4, SETD5, SETD6, SETD7, SETD8, SETD9, SETDB1, SETDB2, SETMAR, SMYD1, SMYD2, SMYD3, SMYD4, SMYD5, SUV39H1, SUV39H2, SUV420H1, or SUV420H2.
The compound inhibits the trimethylation of histone H3 at lysine 4 (H3K4me3) and/or methylation of histone H4 at lysine 5 (H4K5me).
SMYD3 may regulate multiple overlapping MAP kinase pathway proteins. Accordingly, the compound of the present disclosure may modulate myostatin transcription and/or c-Met transcription.
The compound is assumed to inhibit the MEK-ERK mitogen-activated protein-kinase pathway.
The compound may inhibit methylation of a lysine residue on MAP3K2.
The lysine residue may be K260.
The compound may be administered alone or in the form of a pharmaceutical composition in combination with a pharmaceutically acceptable carrier, diluent or excipient. The compounds, while effective themselves, may be typically formulated and administered in the form of their pharmaceutically acceptable salts as these forms are typically more stable, more easily crystallized and have increased solubility.
The compound may, however, typically be used in the form of pharmaceutical compositions which are formulated depending on the desired mode of administration.
A pharmaceutical composition may comprise a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, and a pharmaceutically acceptable excipient. The compositions may be prepared in manners well known in the art.
The amount of compound in the compositions may be such that it is effective to measurably inhibit one or both of methylation of histone H3 at lysine 4 (H3K4me3) and of histone H4 at lysine 5 (H4K5me) in a biological sample or in a patient. The composition may be formulated for administration to a patient in need of such composition. In using the compounds, they may be administered in any form or mode which may make the compound bioavailable. One skilled in the art of preparing formulations can readily select the proper form and mode of administration depending upon the particular characteristics of the compound selected, the condition to be treated, the stage of the condition to be treated and other relevant circumstances.
The term "pharmaceutically acceptable excipient" may refer to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this disclosure may include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene -polyoxypropylene -block polymers, polyethylene glycol or wool fat.
Compositions as defined above may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this disclosure may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
Pharmaceutically acceptable compositions as defined above may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
Pharmaceutical compositions for parenteral injection may comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate. Proper fluidity may be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservative, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of micro-organisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminium monostearate and gelatin.
If desired, and for more effective distribution, the compounds may be incorporated into slow release or targeted delivery systems such as polymer matrices, liposomes, and microspheres.
The injectable formulations may be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
Alternatively, pharmaceutically acceptable compositions as defined above may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
Pharmaceutically acceptable compositions as defined above may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations may be readily prepared for each of these areas or organs.
Topical application for the lower intestinal tract may be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
For topical applications, the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of compounds as defined above may include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutically acceptable compositions may be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers may include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water. For ophthalmic use, the pharmaceutically acceptable compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylaikonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
Pharmaceutically acceptable compositions as defined above may also be administered by nasal aerosol or inhalation. Such compositions may be prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
Most preferably, pharmaceutically acceptable compositions as defined above may be formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions as defined above may be administered without food. In other embodiments, pharmaceutically acceptable compositions as defined above may be administered with food.
The amount of compound that may be combined with the carrier materials to produce a composition in a single dosage form may vary depending upon the host treated, the particular mode of administration. Preferably, the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present disclosure in the composition will also depend upon the particular compound in the composition.
A method of inhibiting SMYD3 in a cell may comprise administering to a cell a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above.
The activity of a compound as an inhibitor may be assayed in vitro, in vivo or in a cell line. In vitro assays may include assays that determine inhibition of either the methylation activity and/or the subsequent functional consequences, or methylation activity of one or both of histone H3 at lysine 4 (H3K4me3) and histone H4 at lysine 5 (H4K5me), or the methylation of a lysine residue on MAP3K2. In the in vitro assay, SMYD3 catalyzes the methylation of the MAP3K2 peptide substrate by transferring a methyl group from SAM to MAP3K2 peptide and further converts the SAM to SAH. The SMYD3 methyltransferase activity is measured based on the amount of SAH produced from the reaction through the use of coupling enzymes that convert the SAH to ATP.
The inhibition of SMYD3 further comprises the inhibition of cell proliferation.
The cell may be in vitro.
The cell may be from a cell line. The cell line may be an immortalized cell line, a genetically modified cell line or a primary cell line.
The cell line may be selected from the group consisting of HepG2, HCT116, A549, HPAF-II, CFPAC-1, HuH7, SNU398, Hep3B, PLC/PRF/5, HuHl, Bel7404, HCCLM3, HLE, SK-HEP-1, Mahlavu, JHH1, JHH2, JHH4, JHH5, JHH7, SNU354, SNU368, SNU387, SNU423, SNU449, SNU739, SNU761, SNU886, MIA PaCa-2 and HEK293.
The cell may be from tissue of a subject.
The cell may be in a subject.
A method of treating a SMYD-3-related disorder may comprise administering to a subject in need of treatment a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above.
The method as disclosed above may further comprise the step of administering an additional therapeutic agent in the subject.
The compound as disclosed above, or a pharmaceutically form or prodrug thereof, or a composition as disclosed above may be for use in therapy.
The use of a compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above, may be in the manufacture of a medicament for treatment of a SMYD3-related disorder.
The medicament may be administered with an additional therapeutic agent, wherein said medicament may be administered in combination or alteration with the additional therapeutic agent.
A compound as disclosed above, or a pharmaceutically acceptable form or prodrug thereof, or a composition as disclosed above, may be for use in the treatment of a SMYD3-related disorder.
The disorder may be cancer, angiogenic disorder or pathological angiogenesis, fibrosis or inflammatory conditions.
The disorder may be lymphoma, cutaneous T-cell lymphoma, follicular lymphoma, or Hodgkin lymphoma, cervical cancer, ovarian cancer, breast cancer, lung cancer, prostate cancer, colorectal cancer, gastric cancer, pancreatic cancer, sarcoma, hepatocellular carcinoma, leukemia or myeloma, retinal angiogenic disease, liver fibrosis, kidney fibrosis, or myelofibrosis.
The compound may be administered with an additional therapeutic agent, wherein said medicament may be administered in combination or alteration with the additional therapeutic agent.
A process for synthesizing the compound as disclosed above, having the following Formula (III), may comprise the steps of:
Figure imgf000063_0001
(III)
(a) contacting an optionally substituted aminobenzoate ester with a compound having following Formula (Va) to form a cyclized product;
Figure imgf000064_0001
wherein R is selected from the group consisting of H, methyl, COOMe and COOEt;
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form the compound of formula (III);
Figure imgf000064_0002
wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
By way of illustration, the compounds, esters, amides, salts and solvates of Formula (III) may be prepared by a process which comprises an initial reaction step (a) between an aminobenzoate and a carbonyl-containing moiety of Formula (Va). This reaction may be carried out in a solvent. It may occur in a high-boiling solvent. The solvent may be selected from the group consisting of toluene, 1,4- dioxane, n-butanol, diphenyl ether, chlorobenzene, carbon tetrachloride, diethylene glycol, diglyme, hexamethylphosphoramide, o-xylene, m-xylene and p-xylene. The reaction temperature may be in a range of about 100 to about 400 °C, or about 150 to about 400 °C, or about 200 to about 400 °C, or about 250 to about 400 °C, or about 300 to about 400 °C, or about 350 to about 400 °C, or about 150 to about 350 °C, or about 150 to about 300 °C, or about 150 to about 250 °C, or about 150 to about 200 °C, or about 150 to about 350 °C, or about 200 to about 300 °C, or about 250 to about 300 °C, e.g. at about 100 °C, at about 150 °C, at about 200 °C, at about 250 °C, at about 300 °C, at about 350 °C, or at about 400 °C. It may be heated in a sealed tube. It may be in a reflux apparatus. It may be heated by an oil bath or a sand bath. It may be heated using microwave irradiation. The reaction time may vary between 30 min to 6 hours. It may vary in a range of about 30 min to 6 hours, or about 1 hour to 6 hours, or about 1.5 hours to 6 hours, or about 2 hours to 6 hours, or about 2.5 hours to 6 hours, or about 3 hours to 6 hours, or about 3.5 hours to 6 hours, or about 4 hours to 6 hours, or about 5 hours to 6 hours, or about 30 min to 5 hours, or about 30 min to 4 hours, or about 30 min to 3 hours, or about 30 min to 2 hours, or about 30 min to 1 hour, e.g. it may be about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours, or about 5 hours, or about 6 hours. After the reaction is complete, that reaction solution may be diluted with a non-polar solvent. The non-polar solvent may be selected from pentane, hexane, heptane, methyl t-butyl ether, petroleum ether and dichloromethane. The product may precipitate out of the solution.
In reaction step (b) the ensuing amino-enone may be treated with a halogenating reagent. The halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride. The reaction may be in a solvent. It may be in a non-polar solvent. It may be in a solvent selected from the group consisting of hexane, cyclohexane, benzene, toluene, 1,4-dioxane, chloroform, diethyl ether and dichloromethane. It may be at an elevated temperature. The temperature may be in a range of about 30 to 120 °C, or about 50 to 120 °C, or about 70 to 120 °C, or about 90 to 120 °C, or about 110 to 120 °C, or about 30 to 100 °C or about 30 to 80 °C, or about 30 to 60 °C, or about 30 to 40 °C or at about 30 °C, or at about 50 °C, or at about 70 °C, or at about 90 °C, or at about 110 °C, or at about 130 °C. The reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours. The reaction product may be purified by aqueous work-up followed by chromatography.
For the hydrolysis of reaction step (c), the ester-containing starting material may be treated with a base in a solvent. The base may be selected from a variety of bases including inorganic bases or nitrogen bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used. For example, the base may be lithium hydroxide. The solvent mixture may contain two solvents. At least one of these solvents may be a polar solvent. The solvent mixture may contain methanol. The solvent mixture may contain 1,4-dioxane. The solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane. The reaction may be followed by an aqueous work-up under acidic conditions. The pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
The selective functionalizing of the carboxylic acid of step (c) may be performed under peptide coupling reaction conditions known to the person skilled in the art. In particular, it may involve a peptide coupling reagent selected from the group consisting of HATU, Ν,Ν'- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride. It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used. A solvent may be used. The solvent may include polar aprotic solvents. The solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane. The reaction may be performed at room temperature. The reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours. The reaction product may be purified by aqueous work-up followed by chromatography.
A process for synthesizing the compound as disclosed above, having the following Formula (III), wherein R1 is optionall a halogen or hydrogen, may comprise the steps of:
Figure imgf000066_0001
(HI),
(a) contacting an optionally substituted aminobenzoate ester with a compound having the following Formula (Vb) and phosphorus ox chloride to form a halogenated cyclized product;
Figure imgf000066_0002
(Vb)
(b) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form an amide; and
O
Fr
NH
R7 R6
(VI)
(c) selectively functionalizing at least one halogen of the halogenated cyclized product of step (a) with a group having the following formula (Vila) or formula (Vllb) under reaction conditions to form the compound of formula (III);
Figure imgf000066_0003
(Vila) (Vllb) wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
By way of illustration, the compounds, esters, amides, salts and solvates of Formula (III) may alternatively be prepared by a process which comprises an initial reaction step (a) between an aminobenzoate and a carbonyl-containing moiety of Formula (Vb). The starting materials may be treated with a halogenating reagent. The halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride. The reaction may occur without a solvent. The temperature for mixing the solvents may be in a range of about -30 to 10 °C, or about - 20 to 10 °C, or about 10 to 10 °C, or about 0 to 10 °C, or about -30 to 0 °C, or about -30 to -10 °C or about -30 to -20 °C, or at about -30 °C, or at about -20 °C, or at about -10 °C, or at about 0 °C, or at about 10 °C. The reaction temperature may be raised to about 60 to 150 °C, or to about 80 to 150°C, or to about 100 to 150°C, or to about 120 to 150°C, or to about 140 to 150°C, or to about 60 to 130°C, or to about 60 to 110°C, or to about 60 to 90°C, or to about 60 to 70°C, or to about 60°C, or to about 80°C, or to about 100°C, or to about 110°C, or to about 130°C, or to about 150°C. The reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours. The reaction product may be purified by aqueous work-up followed by chromatography.
In the hydrolysis of reaction step (b) the ester -containing starting material may be treated with a base in a solvent. The base may be selected from a variety of bases including inorganic bases or nitrogen bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used. For example, the base may be lithium hydroxide. The solvent mixture may contain of two solvents. At least one of these solvents may be a polar solvent. The solvent mixture may contain methanol. The solvent mixture may contain 1,4-dioxane. The solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane. The reaction may be followed by an aqueous work-up under acidic conditions. The pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
The selective functionalizing of the carboxylic acid in step (b) may be performed under peptide coupling reaction conditions known to the person skilled in the art. In particular, it may involve a peptide coupling reagent selected from the group consisting of HATU, Ν,Ν'- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride. It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used. A solvent may be used. The solvent may include polar aprotic solvents. The solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane. The reaction may be performed at room temperature. The reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours. The reaction product may be purified by aqueous work -up followed by chromatography.
Step (c) may be performed under cross coupling reaction conditions known to the person skilled in the art. In particular, it may involve a cross coupling catalyst. The catalyst may be selected from a palladium-containing catalyst. It may be selected from the group consisting of Pd(PPh3)4 or 1, 1 '- [bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with dichlorome thane. It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used. A solvent mixture may be used. The solvent mixture may contain water. The solvent mixture may contain a polar solvent. The solvent mixture may be a mixture of water and 1,4-dioxane. The reaction temperature may be in the range of about 60 to 150 °C, or to about 80 to 150 °C, or to about 100 to 150 °C, or to about 120 to 150 °C, or to about 140 to 150 °C, or to about 60 to 130 °C, or to about 60 to 110 °C, or to about 60 to 90 °C, or to about 60 to 70 °C, or to about 60 °C, or to about 80 °C, or to about 100 °C, or to about 110 °C, or to about 130 °C, or to about 150 °C. The reaction may be performed under conditions known to the person skilled in the art. The reaction product may be purified by filtration followed by chromatography.
A process for synthesizing the compound as disclosed above having the following formula (IV), comprises the ste s of;
Figure imgf000068_0001
(IV)
(a) contacting an amino substituted terephthalic acid or an ester thereof; with an optionally substituted cyclic ketone having the following Formula (VIII) to form a cyclized product;
Figure imgf000068_0002
(VIII)
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) optionally selectively hydrolyzing the ester of step (a) to a carboxylic acid; and (d) selectively functionalizing the carboxylic acid of the cyclized product of step (a) or step (c) with a group having the following formula (VI) under reaction conditions to form the compound of formula (IV);
Figure imgf000069_0001
(VI)
wherein step (b), (c) and (d) may be performed simultaneously, sequentially or in any order.
The reaction steps may be described as disclosed above.
The process comprises the step of optionally hydrolyzing the carboxylic acid ester after formation of the cyclized product in step (a). The hydrolyzing step may be performed under conditions known to the person skilled in the art.
By way of illustration, the compounds, esters, amides, salts and solvates of Formula (IV) may be prepared by a process which comprises an initial reaction step (a) between a terephthalic acid or an ester therof, and a carbonyl-containing moiety of Formula (VIII). This reaction may be carried out in a solvent. It may occur in a high-boiling solvent. The solvent may be selected from the group consisting of toluene, 1,4-dioxane, n-butanol, diphenyl ether, chlorobenzene, carbon tetrachloride, diethylene glycol, diglyme, hexamethylphosphoramide, o-xylene, m-xylene and p-xylene. The reaction temperature may be in a range of about 100 to about 400 °C, or about 150 to about 400 °C, or about 200 to about 400 °C, or about 250 to about 400 °C, or about 300 to about 400 °C, or about 350 to about 400 °C, or about 150 to about 350 °C, or about 150 to about 300 °C, or about 150 to about 250 °C, or about 150 to about 200 °C, or about 150 to about 350 °C, or about 200 to about 300 °C, or about 250 to about 300 °C, e.g. at about 100 °C, at about 150 °C, at about 200 °C, at about 250 °C, at about 300 °C, at about 350 °C, or at about 400 °C. It may be heated in a sealed tube. It may be in a reflux apparatus. It may be heated by an oil bath or a sand bath. It may be heated using microwave irradiation. The reaction time may vary between 30 min to 6 hours. It may vary in a range of about 30 min to 6 hours, or about 1 hour to 6 hours, or about 1.5 hours to 6 hours, or about 2 hours to 6 hours, or about 2.5 hours to 6 hours, or about 3 hours to 6 hours, or about 3.5 hours to 6 hours, or about 4 hours to 6 hours, or about 5 hours to 6 hours, or about 30 min to 5 hours, or about 30 min to 4 hours, or about 30 min to 3 hours, or about 30 min to 2 hours, or about 30 min to 1 hour, e.g. it may be about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours, or about 5 hours, or about 6 hours. After the reaction is complete, that reaction solution may be diluted with a non-polar solvent. The non-polar solvent may be selected from pentane, hexane, heptane, methyl t-butyl ether, petroleum ether and dichloromethane. The product may precipitate out of the solution.
In reaction step (b) the ensuing amino-enone may be treated with a halogenating reagent. The halogenating reagent may be phosphoryl-containing. It may be phosphorous oxychloride. Alternatively, it may be oxalyl chloride. The reaction may be in a solvent. It may be in a non-polar solvent. It may be in a solvent selected from the group consisting of hexane, cyclohexane, benzene, toluene, 1,4-dioxane, chloroform, diethyl ether and dichloromethane. It may be at an elevated temperature. The temperature may be in a range of about 30 to 120 °C, or about 50 to 120 °C, or about 70 to 120 °C, or about 90 to 120 °C, or about 110 to 120 °C, or about 30 to 100 °C or about 30 to 80 °C, or about 30 to 60 °C, or about 30 to 40 °C or at about 30 °C, or at about 50 °C, or at about 70 °C, or at about 90 °C, or at about 110 °C, or at about 130 °C. The reaction time may be between about 30 min and 4 hours, or between about 1 hour and 4 hours, or between about 2 hours and 4 hours, or between about 3 hours and 4 hours, or between about 30 min and 3 hours, or between about 30 min and 2 hours, or between about 30 min and 1 hour, or about 30 min, or about 1 hour, or about 2 hours, or about 3 hours, or about 4 hours. The reaction product may be purified by aqueous work-up followed by chromatography.
For the hydrolysis of reaction step (c), the ester-containing starting material may be treated with a base in a solvent. The base may be selected from a variety of bases including inorganic bases or nitrogen bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide, lithium hydroxide or sodium bicarbonate may be used. For example, the base may be lithium hydroxide. The solvent mixture may contain of two solvents. At least one of these solvents may be a polar solvent. The solvent mixture may contain methanol. The solvent mixture may contain 1,4-dioxane. The solvent mixture may be a mixture, for example, of methanol and 1,4-dioxane. The reaction may be followed by an aqueous work-up under acidic conditions. The pH of the aqueous work-up may be adjusted to about 2, about 3, about 4, or about 5, it may be, for example, 3.
The selective functionalizing of the carboxylic acid of step (d) may be performed under peptide coupling reaction conditions known to the person skilled in the art. In particular, it may involve a peptide coupling reagent selected from the group consisting of HATU, Ν,Ν'- dicyclohexylcarbodiimide, HBTU, hydroxybenzotriazole, propyl phosphonic anhydride and phosphorous oxychloride. It may involve a base selected from the group of nitrogen bases. It may involve bases selected from the group of inorganic bases. For example, triethylamine, pyridine, sodium hydroxide, potassium hydroxide or sodium bicarbonate may be used. A solvent may be used. The solvent may include polar aprotic solvents. The solvent may be selected from the group consisting of tetrahydrofuran, ethyl acetate, acetone, DMF, acetonitrile, dimethylsulfoxide or nitromethane. The reaction may be performed at room temperature. The reaction time may be between about 1 hour and 16 hours, or between about 1 hour and 14 hours, or between about 1 hour and 12 hours, or between about 1 hour and 10 hours, or between about 1 hour and 8 hours, or between about 1 hour and 6 hours, between about 1 hour and 4 hours, between about 1 hour and 2 hours, between about 3 hours and 16 hours, between about 5 hours and 16 hours, between about 6 hours and 16 hours, between about 8 hours and 16 hours, between about 10 hours and 16 hours, between about 12 hours and 16 hours, between about 14 hours and 16 hours, or about 1 hour, or about 2 hours, or about 4 hours, or about 6 hours, or about 8 hours, or about 10 hours, or about 12 hours, or about 14 hours, or about 16 hours. The reaction product may be purified by aqueous work -up followed by chromatography.
For the purposes of the processes described above, the term "at least one ketone" refers to 1, 2 or 3 ketone moieties and the term "at least one halogen" refers to 1, 2 or 3 halogen moieties.
The compounds as defined above may be made according to the general processes as disclosed above or according to the general principles of the working examples. Examples
Non-limiting examples of the disclosure will be further described in greater detail by reference to specific Examples, which should not be construed as in any way limiting the scope of the invention.
Example 1
List of abbreviations used
Names/terms Abbreviations
Dichloromethane DCM
Dimethylformamide (N,N-) DMF
Dimethyl sulfoxide DMSO equivalent equiv
High-performance liquid chromatography or high-pressure liquid chromatography HPLC
Nuclear Magnetic Resonance NMR
Tetrahydrofuran THF
Materials and Methods of the Biological Assays
SMYD3 Biochemical Assay
A SMYD3 enzymatic assay was developed using Promega's Methyltransferase -GloTM reagents. In the assay, SMYD3 catalyzes the methylation of the MAP3K2 peptide substrate by transferring a methyl group from SAM to MAP3K2 peptide and further converts the SAM to SAH. The SMYD3 methyltransferase activity is measured based on the amount of SAH produced from the reaction through the use of coupling enzymes that convert the SAH to ATP. The MTase-Glo detection solution then catalyzes the formation of light from ATP.
For the IC50 determination, the compounds were incubated with 0.4 μΜ of SMYD3 enzyme for 30 min in low volume 384 well plates. A final concentration of 1.0 μΜ and 10 μΜ SAM and MAP3K2 peptide were added and further incubated for 30 min at room temperature before adding the MTase Glo and detection reagent. Reaction signals were detected using microplate readers on luminescent mode (Safire Tecan). The IC50 was determined by non-linear regression, using GraphPad Prism version, 5.03.
Cells and Reagents
The cell proliferation assays were tested in several cell lines, including HepG2, HCT116, A549, HPAF-II, CFPAC-1, HuH7, SNU398, Hep3B, and HEK293. All cell lines are from ATCC. HepG2, HPAF-II and HEK293 are cultured in Eagle's MEM media supplemented with fetal bovine serum. HCT116 is cultured in McCoy media supplemented with fetal bovine serum. Huh-7 is cultured in DMEM low glucose (1000 mg/L glucose) with 10% FBS, 1% L-Glutamate and 1% Penicillin/ Streptomycin. SNU398 is cultured in RPMI with 10% FBS with 1% L-Glutamate and 1% Penicillin/ Streptomycin. Hep3B is cultured in Eagle's MEM with 10% FBS, 1% L- Glutamate and 1% Penicillin/ Streptomycin. A549 is cultured in RPMI media supplemented with fetal bovine serum while CFPAC-1 is cultured in IMDM media supplemented with fetal bovine serum. All media and serum are purchased from Gibco (Lifetech). All cells were grown in a temperature controlled incubator at 37 °C and 5% C02.
Cell Proliferation Assay
Cell proliferation assay was performed using CellTiter-Glo Luminescent Cell Viability Assay (Promega) following manufacturer's instructions. The cell -line of interest was treated with compounds that were serial diluted in its respective media. Plates were incubated for 3 days at 37 °C in 5% C02. After 3 days, an equal volume of Cell Titer Glo reagent was added. Plates were rocked on a rotator for 2 h. 100 μL· of each well were transferred to a 96-well opaque plate, and luminescence emitted was measured with the Tecan Safire II.
Target engagement assay
The target engagement assay was performed with HEK293 that is engineered to overexpress SMYD3 (HEK293-SMYD3). The plasmid SMYD3 (Myc-DDK-tagged-Human SET and MYND domain containing 3) was purchased from Origene (RC230064) and transfected with lipofectamine 2000 (Invitrogen) into HEK293 (ATCC). The cell line is cultured in Eagle's MEM media supplemented with fetal bovine serum and geneticin (Invitrogen). Presence of over -expressed SMYD3 is confirmed with western blot with antibody against SMYD3 and the MYC tag as well as with RT-PCR with SMYD3 primers.
The cells were seeded in 6 well plates. After seeding for 24 h, the cells were treated with either DMSO or 25 μΜ compound and incubated for 24hr. The cells were trypsinized and the lysate was extracted with RIPA buffer (Santa Cruz). The total protein concentration of lysate is quantified using the standard Bradford assay (Biorad protein assay, microplate standard assay).
Western blot analysis
Western blot analysis was performed using antibody against SMYD3 in HEK293 cells over- expressing SMYD3 (HEK293-SMYD3) treated with compound at different time points. 15.0 μg of cell lysate was diluted in 2 x Laemmli sample buffer (Bio-Rad) and boiled at 100 °C on heat block for 5 min. Lysates were separated using NuPAGE® 4-12% Bis-Tris precast polyacrylamide gels (Lifetech) at 200 V, 400 A for 40 min. The electrophoresed protein was transferred onto the nitrocellulose membrane for 7 min using the iBlot 2 Dry Blotting System (Lifetech) . After 1 h incubation in blocking buffer [PBS (phosphate buffered saline) with 0.1% Tween 20 and 5% dry milk], the membrane was probed with anti-SMYD3 primary antibody mouse [GT1088] (Ab 177163, Abeam), 1 :2500 dilution in PBS, 0.1% Tween 20 and 5% dry milk overnight at 4 °C, followed by three washes (15 min each wash) in PBS, 0.1% Tween 20 on the next day. This was further continued with 1 h incubation with peroxidase-conjugated secondary antibody (anti-mouse-HRP, NA9310V (GE)), 1:5000 dilutions in PBS, 0.1% Tween 20 and 5% dry milk followed by three washes (15min each wash) in PBS, 0.1% Tween 20. The nitrocellulose membrane was developed with enhanced Chemiluminescence (ECL) mixture (Amersham, Aylesbury, UK), incubated for 5 min and exposed using FluorChem E System instrument (Protein Simple).
Western blot analysis for detecting methylated MAP3K2 was carried out using a customized Anti- me2/me3-Lys 260 MAP3K2 at 1 :500 and the total MAP3K2 was detected using Anti-MEKK2 (AB33918) using a 1 : 10,000 dilution.
Soft Agar Colony formation assay
Hep G2 cells were purchased from ATCC. Hep G2 cells were maintained in Eagle's Minimum Essential Medium (Sigma, Cat No: #M0643) supplemented with 10% Fetal Bovine Serum (Hyclone, Cat No: SV30087.03), 2 mM L-glutamine, 100 units/mL penicillin and 100 μg/mL streptomycin (Life Technologies, Cat No: 10378-016). The soft agar assays were performed in concordance to the ETC approved method report for soft agar assay (ETC document number: RD0019). Briefly, 600 μΕ of 0.6% agar was added to 24-well plate (Corning, Cat No: 3738) to form the base layer. This is followed by the addition of 500 μΕ of 0.36% agar middle layer (containing 10Ό00 Hep G2 cells). Lastly, 500 μΕ of fresh growth medium (containing the corresponding serially diluted compound) was added above the middle layer. The plates were incubated at 37 °C with 5% carbon dioxide in a humidified incubator for 1 to 2 weeks. 70 μΕ of thiazolyl blue tetrazolium bromide (5 mg/mL, Sigma Cat No: M5655) was added to each well and the plates were incubated at 37 °C for 2 h. Colonies were counted with GelCount® instrument (Oxford Optronix). The colony counts were plotted against compound concentrations using the Graphpad Prism software. In addition, the software was used to perform non-linear curve fitting and the calculation of IC50.
Example 2
SMYD3 biochemical assay
The ability of compounds to inhibit the catalytic function of SMYD3 was tested using the MTase assay by using the MAP3K2 as a peptide substrate. The compounds were found to inhibit the methyltransferase activity of SMYD3. The effect of compounds on the methyltransferase activity of SMYD3 using MAP3K2 peptide as a substrate can be found in Fig. 1 (Compound A066; A074; B019; A088). The data has been summarized in Table 1.
Table 1. Summary of biochemical IC50 of SMYD3 inhibitors.
Compound A066 A074 B019 A088
IC50 ^M) 0.04614 0.1519 0.05951 0.09203 Example 3
SMYD3 compounds inhibit the proliferation of different cancer cells
The anti-proliferative effects of SMYD3 inhibition were explored in different cancer cell lines. The dose-response curves are shown in Fig. 2. All cell lines responded to the SMYD3 inhibitors with GI50 values ranging from 11 to 50 μΜ. A subset of the data is summarized in Table 2.
Table 2. Summary of anti-proliferative activity of SMYD3 inhibitors in different cancer cell lines (Compounds A066; A074; B019; A088).
Figure imgf000074_0001
ND: not determined
Example 4
SMYD3 compounds inhibit the SMYD3 mediated methylation of MAP3K2 in cells
SMYD3 target engagement with B019 was demonstrated in HEK293 cells transiently transfected with Myc-tagged SMYD3. The cells were treated with 25 μΜ of the compound, overnight before analyzing the lysate on Western Blot. A 35-37% reduction in SMYD3 levels was observed using an antibody (Anti-SMYD3 #Abl77163) (1 :5000) against SMYD3, as shown in Fig. 3.
B019 was further tested for its ability to inhibit SMYD3 in cells through its effects on cellular MAP3K2 methylation. This was performed using an antibody against the MAP3K2 (me2/me3), Anti- ME2/ME3-K260-MAP3K2 (1 :500) and total MAP3K2, Anti-MEKK2 #ab33918 (1: 10,000). B019 inhibited the methylation of MAP3K2 in the cells without changing the total MAP3K2 levels. Whereas, the less active compound X4 (please refer to Comparative Example 1) was unable to inhibit the methylation of MAP3K2 at 25 μΜ.
Example 5
Inhibition of anchorage independent growth of cancer cells with SMYD3 inhibitors
Compound A074 and B019 inhibited colony formation in Hep G2 cell line in the soft agar assay (Fig. 4). At the highest concentration tested (10 μΜ), both compounds inhibited about 100% of HepG2 colony formation. Each data point in the dose response curve consists of average colony count from two wells and the error bar is the standard deviation of the count. Fig. 4B shows an IC50 of 0.71 μΜ and a maximum inhibition of 100%, Fig. 4D shows an IC50 of 0.23 μΜ and maximum inhibition of 97.2%.
Example 6
General Reaction Schemes
General Procedures. All reactions were performed using oven-dried round-bottomed flasks or reaction vessels. Where appropriate, reactions were carried out under an inert atmosphere of nitrogen with dry solvents, unless otherwise stated. Dry dichloromethane (DCM), tetrahydrofuran (THF), N,N- dimethylformamide (DMF), dimethylsulfoxide (DMSO), toluene (PhMe), acetonitrile (MeCN) and methanol (MeOH) were purchased at the highest commercial quality. Yields refer to chromatographically and spectroscopically ('H NMR) homogeneous materials, unless otherwise stated. Reagents were purchased at the highest commercial quality and used without further purification, unless otherwise stated. Reactions were monitored by thin-layer chromatography carried out on 0.25 mm E. Merck silica gel plates (60F-254) using ultraviolet light as visualizing agent and potassium permanganate and heat as developing agents. NMR spectra were recorded on a Bruker/ Agilent 400 spectrometer and were calibrated using residual undeuterated solvent as an internal reference (CDC13: !H NMR = 7.26; DMSO-<¾: *H NMR = 2.50; CD3OD: lU NMR = 3.31). The following abbreviations or combinations thereof were used to explain the multiplicities: s = singlet, d = doublet, t = triplet, q = quartet, sex = sextet, m = multiplet, br = broad. Liquid chromatography mass spectra (LCMS) were recorded on an Agilent or Shimadzu mass spectrometer using ESI-TOF (electrospray ionization -time of flight).
1. General rocedure for the synthesis of compounds with general formula (IV).
Figure imgf000075_0001
(IV)
Step 1: General Procedure A
A solution of 2-amino-terephthalic acid (1 equiv) and cyclic ketone (VIII) (1.2 equiv) in diphenyl ether (10 mL/g) was heated to 300 °C for 1-3 h in a sealed tube. Upon cooling to room temperature, the reaction was diluted with hexanes and the resulting solid was collected by filtration to afford cyclized product SI. Cyclized product SI was used without subsequent purification.
Step 2: General Procedure B Cyclized product SI (1 equiv) was treated with phosphorus oxychloride (10 mL/g) and the mixture was heated at 100 °C for 4 h. Upon cooling, the crude mixture was either: (a) concentrated under reduced pressure, diluted with cold water and stirred until a free solid formed. The solid was collected by filtration and washed with hexanes to afford compound S2, or: (b) diluted with dichloromethane and poured into a separating funnel containing cold 2 M aqueous sodium hydroxide. The organic layer was separated and the aqueous layer was extracted thrice with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford chlorinated compound S2.
Step 3: General Procedure C
Condition 1: Amide coupling using l-[bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate (HATU)
Chlorinated compound S2 (1 equiv), amine (VI) (1-2 equiv) and triethylamine (2-4 equiv) were dissolved in N,N-dimethylformamide (0.1 M) and the solution was cooled to 0 °C. HATU (1.5 equiv) was added and the reaction was quenched upon completion based on LCMS analysis (<1 h) by the addition of water. The aqueous layer was extracted 3-5 times with ethyl acetate, and the combined organic layers were washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography to afford compound with general formula (IV).
Condition 2: Amide coupling using propylphosphonic anhydride (T3P)
To a suspension of compound S2 (1 equiv) and amine (VI) (0.5-1.2 equiv) in tetrahydrofuran (5-10 mL/g) at 0 °C was added T3P (50% solution in ethyl acetate) (2-3 equiv) and triethylamine (10 equiv). The resulting mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with ethyl acetate, and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (ethyl acetate/hexanes) or preparative HPLC to afford compound with general formula (IV).
2. General Procedure for reduction of compounds with nitrile group: General Procedure D
Potassium borohydride (4 equiv), Raney nickel (moist weight, approximately 1 equiv) were dissolved in dry ethanol (20 mL). To the resulting slurry was added compound to be reduced (1 equiv) while stirring. After vigorously stirring at room temperature for 45 min, the reaction mixture was filtered. The organic layer was evaporated and residue was dissolved in ethyl acetate. The resulting solution was washed with water and the organic layer was dried over anhydrous sodium sulfate and concentrated. The crude product was purified by flash column chromatography on silica gel (methanol/dichloromethane) followed by preparative-HPLC to get the desired product. 3. Alternative General Procedure for com ounds with general formula (IV).
Figure imgf000077_0001
Step 1: General Procedure E
A solution of 2-amino-4-(methoxycarbonyl)benzoic acid (1 equiv) and cyclic ketone (VIII) (1.2 equiv) in diphenyl ether (10 mL/g) was maintained at 300 °C for 1-3 h in a sealed tube. The reaction mass was cooled to room temperature and diluted with hexanes. The resultant solid was collected by filtration, washed with hexanes and dried to afford cyclized product S3.
Step 2: General Procedure F
To a solution of Compound S3 (1 equiv) in a mixture of tetrahydrofuran, methanol and water (1 : 1 :0.5 mL/g) was added lithium hydroxide monohydrate (3 equiv). The reaction mixture was stirred at room temperature for 2-5 h. The reaction mixture was concentrated under reduced pressure, the residue was diluted with water (20 mL), washed with EtOAc (3 x 50 mL). The aqueous layer was separated and acidified to a pH 2 using aqueous 2 M hydrochloric acid and the resulting solid was collected by filtration to afford compound SI.
Step 3: see General Procedure C for synthesis ofS4.
Step 4: General Procedure G
A mixture of cyclized product S4, phosphorus oxychloride (5 mL/g) in dichloromethane (10 mL/g) was heated at 70 °C for 2 h. Upon cooling, the reaction mass was concentrated under reduced pressure and the reaction mass was diluted with ethyl acetate and washed with saturated sodium bicarbonate solution, brine and dried over anhydrous sodium sulfate, filtered, and concentrated. The crude product was purified by preparative-HPLC to afford compound with general formula (IV).
4. General procedure for the synthesis of compounds with general formula (III).
Figure imgf000078_0001
Step 1: General Procedure H
A solution containing compound (Va) (1 equiv), an optionally substituted aminobenzoate ester (R17 = H, 1.2 equiv) and the hydrochloride salt of the aniline derivative (0.01 equiv) in toluene was heated to reflux in a Dean-Stark apparatus overnight. The solution was cooled to room temperature and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/petroleum ether) to afford the intermediate enamine. This intermediate was dissolved in diphenyl ether (10 mL/g) and was heated to 330 °C for 2-4 h in a sealed tube. The reaction mass was cooled to room temperature and diluted with hexanes. The resulting solid was collected by filtration, washed with hexanes and dried to afford cyclized product S5.
Step 1A: General Procedure I
To a reaction vessel containing compound an optionally substituted aminobenzoate ester (R17 = COOH, 1 equiv) and ketone (Va) (1-2 equiv) was added phosphorus oxychloride (1.6 mL/mmol). The resulting mixture was heated to 100 °C for 3 h before cooling it to 0 °C. The cooled mixture was dissolved in dichloromethane (40 mL/mmol) and then basified by adding 2 M aqueous sodium hydroxide (40 mL/mmol). The organic layer was separated and the aqueous phase was extracted twice more with dichloromethane. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford compound S6 directly.
Step 2: General Procedure J
The solution of compound S5 (1 equiv) in phosphorus oxychloride (30 mL/mmol) was heated at 110 °C for 1 h. The reaction mixture was concentrated under reduced pressure and poured into ice water and the compound was extracted into 10% methanol in dichloromethane (2 x 100 mL). The combined organic layer was washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford compound S6.
Step 3: General Procedure K
To a solution of S6 (1 equiv) in methanol (2 mL/mmol) and 1,4-dioxane (1 mL/mmol) was added an aqueous solution of lithium hydroxide (10 equiv) in water (2 mL/mmol). The mixture was heated to 50 °C for 1 h until all contents were soluble. The solution was cooled to room temperature before ethyl acetate (25 mL) was added. The mixture was acidified to pH 4 with 1 M aqueous hydrochloric acid and then the organic layer was separated. The aqueous layer was extracted thrice with ethyl acetate and the combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford compound S7.
Step 4: see General Procedure C to synthesize compounds with general formula (III). 5. General procedure 2 for synthesis of compounds with general formula (III).
Figure imgf000079_0001
Step 1: Synthesis of intermediate S8.
To a round-bottomed flask containing malonic acid (Vb) (11.36 g, 0.1092 mol, 1.1 equiv) was added phosphorus oxychloride (56 mL, 0.599 mmol, 6 equiv) dropwise at 0 °C. After 30 min, methyl 3- aminobenzoate (15 g, 0.0992 mol) was added portion wise (3 additions) to the cold mixture. After warming to room temperature, the mixture was heated to reflux for 4 h. Upon cooling, the mixture was diluted with dichloromethane and neutralized to pH 14 using 6M aqueous sodium hydroxide. The organic layer was separated and the aqueous layer was extracted 4 more times with dichloromethane. The combined extracts were washed with saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography to afford S8 as a white solid (1.738 g, 7%).
Step 2: Synthesis of intermediate S9.
Intermediate S8 (1.738 g, 6.96 mmol) was dissolved in methanol (21 mL) and 1,4-dioxane (21 mL). A solution of lithium hydroxide (1.7 g, 0.07095 mol, 10.2 equiv) in water (21 mL) was added. After 30 min, the mixture was acidified to pH 3 with concentrated hydrochloric acid. The organic solvents were removed under vacuum and the white residue was collected via filtration and washed with copious amount of water. The white solid was dried in a vacuum oven to afford intermediate S9 (1.549 g, 92%).
Step 3: Synthesis of intermediate S10, See General Procedure C.
Step 4: Synthesis of compounds with general formula (III). General Procedure L.
Intermediate S10 (1 equiv), boronic acid (VII) (1.3 equiv) and potassium phosphate tribasic (3 equiv) were added to a reaction vessel. A mixture of 1 ,4-dioxane/water (4 : 1) was added (0.07 M) and the resulting mixture was degassed with nitrogen. [1,1 -
Bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (0.1 equiv) was added and the resulting mixture was heated to 100 °C (30 min to 12 h). Upon cooling, the mixture was filtered through a pad of Celite® with ethyl acetate, and the mixture was concentrated. The product was purified either with column chromatography or preparative HPLC to afford compound with general formula (III).
Example 7
The following Table 3 shows a list representing the exemplified compounds of this disclosure, together with the biological activity data. The ability of the exemplified compounds to inhibit the catalytic function of SMYD3 was tested using the MTase assay by using the MAP3K2 as a peptide substrate. The compounds were found to inhibit the methyltransferase activity of SMYD3.
Compound List
Table 3. Table listing the structure and IC50 of the compounds disclosed
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Example 8
Characterization data
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbon l)piperazine-l-carboxylate (A001)
Figure imgf000120_0002
Compound A001 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl piperazine- 1-carboxylate as starting materials.
*H NMR (400 MHz, CDC13) δ 8.19 (d, / = 8.6 Hz, 1 H), 7.94 (d, / = 1.2 Hz, 1 H), 7.65 (dd, / = 8.6, 1.5 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.80-3.28 (m, 8 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 1.90 (s, 4 H), 1.66-1.47 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 416.1 [M + H+] with a purity of >95%.
Allyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbon l)piperazine- 1-carboxylate (A002)
Figure imgf000120_0003
A002 Compound A002 was prepared according to General Procedure CI, step 3a, using commercially available 9-chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and allyl piperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, CD3OD) δ 8.41 (d, / = 8.6 Hz, 1 H), 8.02 (d, / = 1.1 Hz, 1 H), 7.76 (dd, / = 8.7, 1.5 Hz, 1 H), 6.05-5.82 (m, 1 H), 5.31 (d, / = 17.6 Hz, 1 H), 5.21 (d, / = 10.4 Hz, 1 H), 4.61 (d, / = 5.5 Hz, 2 H), 3.99-3.44 (m, 2 H), 3.20 (s, 2 H), 3.11 (s, 2 H), 2.02 (dd, / = 6.3, 3.0 Hz, 4 H).
LCMS (ESI-TOF) m/z 414.1 [M + H+] with a purity of >95%. l-(4-(9-Chloro-5,6,7,8-tetrahydroacridine-3-carbon l)piperazin-l-yl)pentan-l-one (A003)
Figure imgf000121_0001
Compound A003 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and valeric acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.22 (d, / = 8.6 Hz, 1 H), 7.97 (d, / = 1.2 Hz, 1 H), 7.68 (dd, / = 8.6, 1.5 Hz, 1 H), 3.83-3.45 (m, 8 H), 3.08 (s, 2 H), 3.00 (s, 2 H), 2.34 (br s, 2 H), 1.91 (s, 4 H), 1.54- 1.43 (m, 2 H), 1.40-1.25 (m, 2 H), 0.88 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 414.1 [M + H+] with a purity of >96%.
Propyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8-diazabicyclo[3.2.1]octane-3- carboxylate (A004)
Figure imgf000121_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,8-diazabicyclo[3.2.1]octane-3- carboxylate to give teri-butyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8- diazabicyclo[3.2.1]octane-3-carboxylate.
Step 2: The above product (190 mg, 0.417 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (8 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,8-diazabicyclo[3.2.1]octan- 8 -yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
Step 3: The crude material from above (72 mg, 0.202 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (56 μL·, 0.405 mmol, 2 equiv) followed by n-propyl chloroformate (34 μΕ, 0.303 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A004 as a white solid (10.0 mg, 11%) upon lyophilization.
*H NMR (400 MHz, CDC13) δ 8.24 (d, / = 8.6 Hz, 1 H), 8.04 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.6, 1.4 Hz, 1 H), 4.88 (s, 1 H), 4.24-3.65 (m, 5 H), 3.40-2.93 (m, 6 H), 2.13-1.89 (m, 5 H), 1.81 (s, 2 H), 1.66 (d, / = 6.2 Hz, 2 H), 1.01-0.76 (m, 4 H).
LCMS (ESI-TOF) m/z 442.2 [M + H+] with a purity of >99%.
Propyl 5-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5-diazabicyclo[2.2.2]octane-2- carboxylate (A005)
Figure imgf000122_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,5-diazabicyclo[2.2.2]octane-2- carboxylate to afford teri-butyl 5-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5- diazabicyclo[2.2.2]octane-2-carboxylate.
Step 2: The above intermediate (120 mg, 0.263 mmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (8 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (2,5-diazabicyclo[2.2.2]octan- 2-yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
Step 3: The crude material (70 mg, 0.197 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (39.8 mg, 0.393 mmol, 2 equiv) followed by n-propyl chloroformate (36.2 mg, 0.295 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A005 as a white solid (10.0 mg, 11%) upon lyophilization.
*H NMR (400 MHz, CDC13) δ 8.24 (d, / = 8.3 Hz, 1 H), 8.05-7.91 (m, 1 H), 7.69-7.54 (m, 1 H), 4.90, 4.54 and 4.42 (multiple peaks, 1 H), 4.28-3.89 (m, 3 H), 3.85-3.64 (m, 2 H), 3.64-3.34 (m, 2 H), 3.14 (s, 2 H), 3.04-2.87 (m, 2 H), 2.24-1.84 (m, 6 H), 1.84-1.62 (m, 2 H), 1.09-0.72 (m, 5 H).
LCMS (ESI-TOF) m/z 442.1 [M + H+] with a purity of >99%.
(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(4-(5-cyclopropylisoxazole-3-carbonyl)piperazin-l- yl)methanone (A006)
Figure imgf000123_0001
Compound A006 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-cyclopropylisoxazole-3-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO) δ 8.22 (d, / = 8.5 Hz, 1 H), 7.99 (s, 1 H), 7.69 (d, / = 8.5 Hz, 1 H), 6.44 (s, 1 H), 3.71 (s, 8 H), 3.08 (s, 2 H), 3.00 (s, 2 H), 2.20 (s, 1 H), 1.91 (s, 4 H), 1.09 (s, 2 H), 0.94 (s, 2 H).
LCMS (ESI-TOF) nt/z 465.1 [M + H+] with a purity of >95%.
(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(4-(5-isobutylisoxazole-3-carbonyl)piperazin-l- yl)methanone (A007)
Figure imgf000123_0002
Compound A007 was prepared according to General Procedure CI using (9-chloro-5,6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-isobutylisoxazole-3-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.22 (d, / = 8.6 Hz, 1 H), 7.99 (s, 1 H), 7.69 (d, / = 8.6 Hz, 1 H), 6.52 (s, 1 H), 3.72 (br s, 8 H), 3.07 (s, 2 H), 3.00 (s, 2 H), 2.70 (s, 2 H), 2.00 (br s, 1 H), 1.91 (s, 4 H), 0.93 (d, / = 6.1 Hz, 6 H).
LCMS (ESI-TOF) nt/z 481.2 [M + H+] with a purity of >97%.
4-(9-chloro-7-methyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000123_0003
Compound A008 was prepared according to General Procedure A, B and C2 using 4- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (d, / =8.8, 1.6 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.31 (m, 8 H), 3.26-3.07 (m, 3 H), 2.47-2.46 (m, 1 H), 2.00-1.97 (m, 2 H), 1.60-1.52 (m, 3 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 430.2 [M + H+] with a purity of >96%.
Propyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8-diazabicyclo[3.2.1]octane-8- carboxylate (A009)
Figure imgf000124_0001
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,8-diazabicyclo[3.2.1]octane-8- carboxylate to give teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate.
Step 2: The resulting intermediate (220 mg, 0.482 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,8-diazabicyclo[3.2.1]octan- 3 -yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
Step 3: The crude material from above (190 mg, 0.534 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.09 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 0.82 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A009 as a white solid (100 mg, 42%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.64 (dd, / = 8.6, 1.5 Hz, 1 H), 4.47-4.23 (m, 2 H), 4.09 (s, 1 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.45-3.33 (m, 2 H), 3.09-2.93 (m, 5 H), 1.90-1.71 (m, 7 H), 1.59 (dd, / = 13.8, 6.7 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 442.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-methylpiperazine-l-carboxylate (A010)
Figure imgf000124_0002
Compound A010 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and (S)-n-propyl 3-methylpiperazine-l- carboxylate as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.63 (dd, / = 8.6, 1.5 Hz, 1 H), 4.33-3.64 (m, 6 H), 3.25-2.85 (m, 7 H), 1.90 (s, 4 H), 1.58 (dq, / = 14.3, 7.1 Hz, 2 H), 1.16 (d, / = 4.7 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.1 [M + H+] with a purity of >96%.
Propyl ( ?)-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-methylpiperazine-l- carboxylate (A011)
Figure imgf000125_0001
Compound A011 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and (R)-n-propyl 3-methylpiperazine-l- carboxylate as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.63 (dd, / = 8.6, 1.5 Hz, 1 H), 4.34-3.66 (m, 6 H), 3.26-2.84 (m, 7 H), 1.90 (s, 4 H), 1.63-1.49 (m, 2 H), 1.16 (d, / = 5.1 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.1 [M + H+] with a purity of >97%.
(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(4-(5-methylisoxazole-3-carbonyl)piperazin-l- yl)methanone (A012)
Figure imgf000125_0002
Compound A012 was prepared according to General Procedure CI using (9-chloro-5, 6,7,8- tetrahydroacridin-3-yl)(piperazin-l-yl)methanone and 5-methylisoxazole-3-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.22 (d, / = 8.8 Hz, 1 H), 7.99 (s, 1 H), 7.70 (d, / = 9.9 Hz, 1 H), 6.48 (s, 1 H), 3.71 (br s, 8 H), 3.08 (s, 2 H), 3.00 (s, 2 H), 2.46 (s, 3 H), 1.91 (s, 4 H).
LCMS (ESI-TOF) m/z 439.1 [M + H+] with a purity of >96%.
Propyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-9- carboxylate (A013)
Figure imgf000126_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 7, 9-diazabicyclo[3.3.1]nonane-9- carboxylate to afford teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9- diazabicyclo[3.3.1]nonane-9-carboxylate.
Step 2: The resulting intermediate (230 mg, 0.489 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (3,9-diazabicyclo[3.3.1]nonan-3- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone.
Step 3: The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A013 as a white solid (50 mg, 41%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.92 and 7.86 (2 x s, 1 H), 7.64 and 7.59 (2 x d, / = 9.1 and 8.6 Hz, 1 H), 4.74-3.71 (m, 6 H), 3.45-3.16 (m, 2 H), 3.04 (s, 2 H), 2.99 (s, 2 H), 2.08 (dt, / = 18.8, 9.7 Hz, 1 H), 1.91 (s, 4 H), 1.79-1.50 (m, 7 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 456.2 [M + H+] with a purity of >98%.
Propyl 6-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6-diazabicyclo[3.1.1]heptane-3- carboxylate (A014)
Figure imgf000126_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,6-diazabicyclo[3.1.1]heptane-3- carboxylate to give teri-butyl 6-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6- diazabicyclo[3.1.1 ]heptane-3-carboxylate
Step 2: The resulting intermediate (200 mg, 0.453 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (3,6-diazabicyclo[3.1.1]heptan-6- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone. Step 3: The crude material from above (100 mg, 0.293 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.99 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A014 as a white solid (50 mg, 40%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.20 (d, / = 8.7 Hz, 1 H), 8.14 (d, / = 1.3 Hz, 1 H), 7.82 (d, / = 8.7 Hz, 1 H), 4.71 (s, 1 H), 4.51 (s, 1 H), 4.15-3.83 (m, 3 H), 3.54-3.18 (m, 2 H), 3.07 (s, 2 H), 2.99 (s, 2 H), 2.80 (dd, / = 14.7, 6.8 Hz, 1 H), 1.90 (s, 4 H), 1.69-1.38 (m, 3 H), 0.98-0.66 (m, 4 H).
LCMS (ESI-TOF) m/z 428.1 [M + H+] with a purity of >95%.
Propyl 9-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-3- carboxylate (A015)
Figure imgf000127_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,9-diazabicyclo[3.3.1]nonane-3- carboxylate to afford teri-butyl 9-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9- diazabicyclo [3.3.1 ] nonane-3 -carboxylate.
Step 2: The resulting intermediate (200 mg, 0.426 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude (3,9-diazabicyclo[3.3.1]nonan- 9-yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone .
Step 3: The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A015 as a white solid (50 mg, 41%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.95 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.6, 1.4 Hz, 1 H), 4.66 (s, 1 H), 4.16-3.85 (m, 4 H), 3.70 (s, 1 H), 3.24-3.10 (m, 2 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 1.97-1.69 (m, 8 H), 1.68-1.49 (m, 4 H), 0.89 (s, 3 H).
LCMS (ESI-TOF) m/z 456.2 [M + H+] with a purity of >95%. 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-ethylpiperazine-l-carboxylate
Figure imgf000128_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-ethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-ethylpiperazine-l-carboxylate
Step 2: The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (2-ethylpiperazin- 1 -yl)methanone .
Step 3: The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A016 as a white solid (50 mg, 40%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.90 (s, 1 H), 7.62 (dd, / = 8.6, 1.4 Hz, 1 H), 4.70-4.21 (m, 1 H), 4.10-3.74 (m, 4 H), 3.67-3.34 (m, 1 H), 3.26-2.79 (m, 7 H), 1.90 (s, 4 H), 1.76-1.38 (m, 4 H), 1.09-0.50 (m, 6 H).
LCMS (ESI-TOF) m/z 444.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5-dimethylpiperazine-l-carboxylate (A017)
Figure imgf000128_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,5-dimethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,5-dimethylpiperazine-l- carboxylate
Step 2: The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,5-dimethylpiperazin- 1 -yl)methanone.
Step 3: The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A017 as a white solid (50 mg, 40%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.24-8.13 (m, 1 H), 7.90 (d, / = 30.2 Hz, 1 H), 7.63 (dd, / = 28.3, 8.5 Hz, 1 H), 4.79 and 4.39 (2 x s, 1 H), 4.29-3.88 (m, 3 H), 3.88-3.63 (m, 1 H), 3.63-3.44 (m, 1 H), 3.37-3.17 (m, 2 H), 3.06 (s, 2 H), 2.98 (s, 2 H), 1.90 (s, 4 H), 1.58 (d, / = 6.9 Hz, 2 H), 1.30-0.97 (m, 6 H), 0.97-0.77 (m, 3 H).
LCMS (ESI-TOF) m/z 444.2 [M + H+] with a purity of >96%.
4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-methylpiperazine-l-carboxylate
Figure imgf000129_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-methylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-methylpiperazine-l-carboxylate.
Step 2: The resulting intermediate (160 mg, 0.36 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(3-methylpiperazin-l-yl)methanone.
Step 3: The crude material from above (100 mg, 0.291 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.00 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.50 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A018 as a white solid (50 mg, 40%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾) δ 8.20 (d, / = 8.6 Hz, 1 H), 7.92 (s, 1 H), 7.65 (d, / = 5.5 Hz, 1 H), 4.58-4.04 (m, 2 H), 3.97 (tt, / = 10.6, 5.3 Hz, 2 H), 3.91-3.32 (m, 2 H), 3.23-2.85 (m, 7 H), 1.89 (t, / = 2.8 Hz, 4 H), 1.65-1.48 (m, 2 H), 1.25-0.93 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.1 [M + H+] with a purity of >97%. Propyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6-diazabicyclo[3.1.1]heptane-6- carboxylate (A019)
Figure imgf000130_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl-3,6-diazabicyclo[3.1.1]heptane-6- carboxylate to give teri-butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,6- diazabicyclo[3.1.1]heptane-6-carboxylate.
Step 2: The resulting intermediate (160 mg, 0.362 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.to afford (3,6-diazabicyclo[3.1.1]heptan-3- yl) (9-chloro-5 ,6,7,8 -tetrahydroacridin-3 -yl)methanone.
Step 3: The crude material from above (100 mg, 0.293 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.99 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.49 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A019 as a white solid (50 mg, 40%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾) δ 8.20 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.61 (d, / = 8.4 Hz, 1 H), 4.26 (s, 1 H), 4.20-3.90 (m, 4 H), 3.76 (s, 1 H), 3.62 (d, / = 13.1 Hz, 1 H), 3.51 (s, 1 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 2.60-2.54 (m, 1 H), 1.90 (s, 4 H), 1.56 (d, / = 8.8 Hz, 3 H), 0.85 (s, 3 H).
LCMS (ESI-TOF) m/z 428.1 [M + H+] with a purity of >95%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,6-dimethylpiperazine-l-carboxylate (A020)
Figure imgf000130_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,6-dimethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,6-dimethylpiperazine-l- carboxylate. Step 2: The resulting intermediate (160 mg, 0.349 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (3 , 5 -dimethylpiper azin- 1 -yl)methanone .
Step 3: The crude material from above (100 mg, 0.279 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.08 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.56 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A020 as a white solid (20 mg, 16%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.6 Hz, 1 H), 7.94 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.6, 1.6 Hz, 1 H), 4.48-4.11 (m, 2 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.41 (s, 2 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 1.90 (s, 4 H), 1.66-1.50 (m, 2 H), 1.32-0.93 (m, 6 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z AAA. I [M + H+] with a purity of >95%.
Isobutyl 4-(9-chloro-5,6,7,8-tetrah droacridine-3-carbonyl)piperazine-l-carboxylate (A021)
Figure imgf000131_0001
To a solution of (9-chloro-5,6,7,8-tetrahydroacridin-3-yl)(piperazin-l-yl)methanone (66 mg, 0.2 mmol) in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (0.35 mL, 2.0 mmol, 10 equiv) and isobutyl chloroformate (0.13 mL, 1.0 mmol, 5 equiv). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A021 (4.8 mg, 6%) as a yellow oil.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.6 Hz, 1 H), 7.96 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.6, 1.6 Hz, 1 H), 3.82 (d, / = 6.5 Hz, 2 H), 3.76-3.44 (m, 8 H), 3.07 (s, 2 H), 3.00 (s, 2 H), 1.91 (s, 5 H), 0.90 (d, / = 6.6 Hz, 6 H).
LCMS (ESI-TOF) m/z 430.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-6-methyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate (A022)
Figure imgf000132_0001
Compound A022 was prepared according to General Procedure A, B and C2 using 3- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.4, 1.6 Hz, 1 H), 3.96 (t, / = 6.4 Hz, 2 H), 3.66-3.21 (m, 8 H), 3.15-3.11 (m, 2 H), 2.94-2.87 (m, 1 H), 2.72-2.65 (m, 1 H), 2.00 (br s, 2 H), 1.60-1.48 (m, 3 H), 1.00 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.8 [M + H+] with a purity of >98%.
4-(9-chloro-8-methyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000132_0002
Compound A023 was prepared according to General Procedure A, B and C2 using 3- methylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.0 Hz, 1 H), 7.93 (s, 1 H), 7.65 (dd, / = 8.4, 1.6 Hz, 1 H), 3.96 (t, / = 6.4 Hz, 2 H), 3.66-3.18 (m, 9 H), 3.17-3.03 (m, 1 H), 3.01-2.93 (m, 1 H), 2.06-1.99 (m, 4 H), 1.60-1.55 (m, 2 H), 1.28 (d, / = 6.8 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.8 [M + H+] with a purity of >97%.
4-(9-chloro-6-phenyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000132_0003
A024
Compound A024 was prepared according to General Procedure A, B, and C2 using 3- phenylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.22 (d, / = 8.8 Hz, 1 H), 7.98 (s, 1 H), 7.68 (dd, / = 8.4, 1.6 Hz, 1 H), 7.38-7.33 (m, 4 H), 7.27-7.23 (m, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.28 (m, 8 H), 3.26-3.18 (m, 4 H), 3.09-3.02 (m, 1 H), 2.22-2.19 (m, 1 H), 2.08-2.04 (m, 1 H), 1.69-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.2 [M + H+] with a purity of >98%.
4-(9-chloro-6-cyano-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000133_0001
Compound A025 was prepared according to the General Procedure A, B, and C2 using 2-(3- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.23 (d, / = 8.0 Hz, 1 H), 7.99 (d, / = 0.8 Hz, 1 H), 7.70 (dd, / = 1.2, 8.4 Hz, 1 H), 3.97 (t, / = 6.8 Hz, 2 H), 3.75-3.28 (m, 11 H), 3.09 (t, / = 6.6 Hz, 2 H), 2.30-2.15 (m, 2 H), 1.65-1.50 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 441.2 [M + H+] with a purity of >96%.
4-(9-chloro-8-cyano-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000133_0002
Compound A026 was prepared according to General Procedure A, B and C2 using 3- oxocyclohexanecarbonitrile (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.28 (d, / = 8.8 Hz, 1 H), 8.01 (d, / = 0.8 Hz, 1 H), 7.73 (dd, / = 1.2, 8.4 Hz, 1 H), 4.77 (d, / = 2.8 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.75-3.00 (m, 10 H), 2.40-1.90 (m, 4 H), 1.65-1.50 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 441.2 [M + H+] with a purity of >95%.
Propyl 4-(9-chloro-6-(pyridin-3-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A027)
Figure imgf000133_0003
A027 Compound A027 was prepared according to General Procedure A, B, and C2 using 3-(pyridin-3- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.62 (br s, 1 H), 8.48 (br s, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 7.98 (s, 1 H), 7.80 (d, / = 6.4 Hz, 1 H), 7.68 (d, / = 8.4 Hz, 1 H), 7.39 (br s, 1 H), 3.97 (br s, 2 H), 3.67-3.40 (m, 8 H), 3.27-3.22 (m, 4 H), 3.05 (br s, 1 H), 2.22 (br s, 1 H), 2.09 (br s, 1 H), 1.59-1.57 (d, / = 6.4Hz, 2 H), 0.88 (br s, 3 H).
LCMS (ESI-TOF) m/z 493.2 [M + H+] with a purity of >95%.
Propyl 4-(6-(aminomethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A028)
Figure imgf000134_0001
Compound A028 was prepared from A025 as starting material following General Procedure D.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.8, 1.6 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.71-3.20 (m, 8 H), 3.21-3.11 (m, 2 H), 2.91-2.82 (m, 1 H), 2.74-2.66 (m, 1 H), 2.62-2.61 (m, 2 H), 2.10 (br s, 1 H), 1.92 (br s, 1 H), 1.61-1.47 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 445.2 [M + H+] with a purity of >96%.
Methyl 9-chloro-6-(4-(propoxycarbonyl)piperazine-l-carbonyl)-l,2,3,4-tetrahydroacridine-2- carboxylate (A029)
Figure imgf000134_0002
Compound A029 was prepared according to General Procedure A, B and C2 using methyl 4- oxocyclohexanecarboxylate (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.8 Hz, 1 H), 7.95 (s, 1 H), 7.67 (d, / = 8.8 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.68-3.60 (m, 5 H), 3.50-3.32 (m, 6 H), 3.27 (br s, 1 H), 3.14-3.08 (m, 4 H), 2.27-2.24 (m, 1 H), 2.01-1.98 (m, 1 H), 1.60-1.55 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) m/z 474.27 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-ethylpiperazine-l-carboxylate (A030)
Figure imgf000135_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-ethylpiperazine-l-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-ethylpiperazine-l-carboxylate.
Step 2: The resulting intermediate (220 mg, 0.48 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (3 -ethylpiperazin- 1 -yl)methanone .
Step 3: The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A030 as a white solid (50 mg, 34%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.92 (s, 1 H), 7.64 (d, / = 7.9 Hz, 1 H), 4.45 (s, 1 H), 4.25-3.70 (m, 4 H), 3.59-3.36 (m, 1 H), 3.22-2.82 (m, 7 H), 1.90 (s, 4 H), 1.78-1.47 (m, 4 H), 0.89 (t, / = 7.3 Hz, 5 H), 0.56 (s, 1 H).
LCMS (ESI-TOF) m/z 444.1 [M + H+] with a purity of >98%.
4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-methylpiperazine-l-carboxylate
Figure imgf000135_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-ethylpiperazine-l-carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-methylpiperazine-l- carboxylate.
Step 2: The resulting intermediate (220 mg, 0.496 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (2-methylpiperazin- 1 -yl)methanone . Step 3: The crude material from above (120 mg, 0.349 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.25 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A031 as a white solid (60 mg, 40%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.63 (dd, / = 8.6, 1.5 Hz, 1 H), 4.96-3.69 (m, 6 H), 3.25-2.78 (m, 7 H), 1.90 (s, 4 H), 1.63-1.45 (m, 2 H), 1.16 (d, / = 4.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(hydroxymethyl)piperazine-l- carboxylate (A032)
Figure imgf000136_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl-2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: The resulting intermediate (170 mg, 0.37 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (3 -(hydroxymethyl)piper azin- 1 -yl)methanone .
Step 3: The crude material from above (120 mg, 0.333 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.74 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.31 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A032 as a white solid (20 mg, 13%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾) δ 8.18 (d, / = 8.6 Hz, 1 H), 7.93 (d, / = 1.2 Hz, 1 H), 7.64 (d, / = 8.6 Hz, 1 H), 4.97-4.63 (m, 1 H), 4.60-4.27 (m, 1 H), 4.26-3.34 (m, 8 H), 3.23-2.91 (m, 6 H), 1.90 (s, 4 H), 1.66-1.49 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 446.1 [M + H+] with a purity of >96%. Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,3-dimethylpiperazine-l-carboxylate (A033)
Figure imgf000137_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,3-dimethylpiperazine-l-carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,3-dimethylpiperazine-l - carboxylate.
Step 2: The resulting intermediate (170 mg, 0.371 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,3-dimethylpiperazin- 1 -yl)methanone.
Step 3: The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A033 as a white solid (30 mg, 20%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.92 (d, / = 1.2 Hz, 1 H), 7.63 (dd, / = 8.6, 1.5 Hz, 1 H), 4.23-4.09 (m, 1 H), 4.08-3.89 (m, 3 H), 3.73-3.60 (m, 1 H), 3.57-3.44 (m, 2 H), 3.43-3.34 (m, 1 H), 3.06 (s, 2 H), 2.98 (s, 2 H), 1.90 (s, 4 H), 1.68-1.50 (m, 2 H), 1.25 (dd, / = 6.9, 2.5 Hz, 6 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 444.1 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-7-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1 -carboxylate (A034)
Figure imgf000137_0002
Compound A034 was prepared according to General Procedure A, B and C2 using methyl 2-(4- oxocyclohexyl)acetate (General Procedure A) and n-propyl piperazine-1 -carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.95 (s, 1 H), 7.65 (dd, / = 8.8, 1.6 Hz 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.65-3.60 (m, 5 H), 3.47-3.26 (m, 6 H), 3.23-3.16 (m, 1 H), 3.15-3.08 (m, 2 H), 2.66-2.50 (m, 3 H), 2.32-2.29 (m, 1 H), 2.03(d, / = 10.0 Hz, 1 H), 1.66-1.55(m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 488.3 [M + H+] with a purity of >95%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,2-dimethylpiperazine-l-carboxylate (A035)
Figure imgf000138_0001
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2,2-dimethylpiperazine-l -carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2,2-dimethylpiperazine-l - carboxylate.
Step 2: The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(3,3-dimethylpiperazin-l-yl)methanone trifluoroacetate salt.
Step 3: The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A035 (3%) as a yellow oil.
LCMS (ESI-TOF) nt/z 444.2 [M + H+].
Propyl 7-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7-diazaspiro[2.5]octane-4- carboxylate (A036)
Figure imgf000138_0002
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 4,7-diazaspiro[2.5]octane-4- carboxylate to afford teri-butyl 7-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7- diazaspiro[2.5]octane-4-carboxylate.
Step 2: The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(4,7-diazaspiro[2.5]octan-7-yl)methanone trifluoroacetate salt. Step 3: The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A036 (3%) as a yellow oil.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.6 Hz, 1 H), 8.00 (s, 1 H), 7.69 (dd, / = 8.6, 1.5 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.79-3.46 (m, 7 H), 3.07 (s, 2 H), 2.99 (s, 2 H), 1.90 (s, 4 H), 1.58 (dd, / = 14.1, 7.1 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H), 0.73 (br s, 4 H).
LCMS (ESI-TOF) m/z 442.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7-diazaspiro[2.5]octane-7- carboxylate (A037)
Figure imgf000139_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 4,7-diazaspiro[2.5]octane-7- carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-4,7- diazaspiro[2.5]octane-7-carboxylate.
Step 2: The crude intermediate was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h, then concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3- yl)(4,7-diazaspiro[2.5]octan-4-yl)methanone trifluoroacetate salt.
Step 3: The crude material from above was dissolved in N,N-dimethylformamide (10 mL) at 4 °C was added N,N-diisopropylethylamine (excess) and propyl chloroformate (excess). The resulting mixture was stirred for 2 h at 4 °C followed by 4 h at room temperature. The mixture was quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to afford A037 (4%) as a yellow oil.
*H NMR (400 MHz, DMSO-<¾) δ 8.20 (d, / = 8.7 Hz, 1 H), 7.92 (s, 1 H), 7.64 (s, 1 H), 3.99 (t, / = 6.4 Hz, 2 H), 3.83-3.15 (m, 6 H), 3.07 (s, 2 H), 2.99 (s, 2 H), 1.90 (s, 4 H), 1.64-1.53 (m, 2 H), 1.07- 0.76 (m, 6 H), 0.58 (s, 1 H).
LCMS (ESI-TOF) m/z 442.1 [M + H+] with a purity of >94%.
Butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate (A038)
Figure imgf000140_0001
Intermediate (9-chloro-5,6,7,8-tetrahydroacridin-3-yl)(piperazin-l-yl)methanone was subjected to General Procedure CI with butyl chloroformate to obtain A038.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.5 Hz, 1 H), 7.95 (d, / = 1.3 Hz, 1 H), 7.66 (dd, / = 8.5, 1.5 Hz, 1 H), 4.03 (t, / = 6.5 Hz, 2 H), 3.83-3.54 (m, 8 H), 3.07 (s, 2 H), 3.00 (s, 2 H), 1.90 (s, 4 H), 1.62-1.50 (m, 2 H), 1.43-1.27 (m, 2 H), 0.90 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.2 [M + H+] with a purity of >96%.
(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(9-(5-methylisoxazole-3-carbonyl)-3,9- diazabicyclo[3.3.1]nonan-3-yl)methanone (A039)
Figure imgf000140_0002
Intermediate (3,9-diazabicyclo[3.3.1]nonan-3-yl)(9-chloro-5,6,7,8-tetrahydroacridin-3-yl)methanone from synthesis of A013 was subjected to General Procedure CI with 5-methylisoxazole-3-carboxylic acid to afford A039.
*H NMR (400 MHz, DMSO-<¾ δ 8.26 (t, / = 8.4 Hz, 1 H), 7.93 (s, 1 H), 7.68 (d, / = 8.4 Hz, 1 H), 6.50 (d, / = 11.2 Hz, 1 H), 4.86-4.46 (m, 3 H), 3.80-3.48 (m, 2 H), 3.23-3.14 (m, 1 H), 3.09 (s, 2 H), 3.00 (s, 2 H), 2.46 (d, / = 19.4 Hz, 3 H), 2.11 (s, 1 H), 2.01-1.53 (m, 9 H).
LCMS (ESI-TOF) m/z 479.2 [M + H+] with purity >96%.
(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(9-(5-methyl-lH-pyrazole-3-carbonyl)-3,9- diazabicyclo[3.3.1]nonan-3-yl)methanone (A040)
Figure imgf000140_0003
A040
Intermediate (3,9-diazabicyclo[3.3.1]nonan-3-yl)(9-chloro-5,6,7,8-tetrahydroacridin-3-yl)methanone from synthesis of A013 was subjected to General Procedure CI with 5-methyl-lH-pyrazole-3- carboxylic acid to afford A040.
lU NMR (400 MHz, DMSO-<¾ δ 8.24 (t, / = 7.8 Hz, 1 H), 7.91 (s, 1 H), 7.66 (d, / = 8.6 Hz, 1 H), 6.33 (d, / = 9.2 Hz, 1 H), 5.22-4.46 (m, 3 H), 3.82-3.57 (m, 3 H), 3.08 (s, 2 H), 3.00 (s, 2 H), 2.24 (d, / = 16.3 Hz, 3 H), 2.17-2.03 (m, 1 H), 1.99-1.48 (m, 8 H).
LCMS (ESI-TOF) m/z 478.2 [M + H+] with purity >94%. Butyl 3-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-9- carboxylate (A041)
Figure imgf000141_0001
Intermediate (3,9-diazabicyclo[3.3.1]nonan-3-yl)(9-chloro-5,6,7,8-tetrahydroacridin-3-yl)methanone from synthesis of A013 was subjected to General Procedure CI with butyl chloroformate as reagent to afford A041.
*H NMR (400 MHz, DMSO-<¾) δ 8.24 (d, / = 8.6 Hz, 1 H), 7.89 (s, 1 H), 7.64 (d, / = 8.6 Hz, 1 H), 4.61 (d, / = 13.1 Hz, 1 H), 4.24 (s, 1 H), 4.16-3.89 (m, 3 H), 3.73-3.55 (m, 2 H), 3.17-3.03 (m, 3 H), 2.99 (s, 2 H), 2.13-1.97 (m, 1 H), 1.99-1.26 (m, 13 H), 1.00-0.75 (m, 3 H).
LCMS (ESI-TOF) m/z 470.3 [M + H+] with purity >96%.
2-Methyl 1-propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l,2- dicarboxylate (A042)
Figure imgf000141_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl methyl piperazine-l,2-dicarboxylate to afford l-(teri-butyl) 2-methyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l,2- dicarboxylate.
Step 2: To a solution of the the above intermediate (184.2 mg, 0.378 mmol) in dichloromethane (1.1 mL) was added trifluoroacetic acid (0.59 mL, 7.71 mmol, 20 equiv). The resulting mixture was stirred for 2 h before concentrating under reduced pressure to give methyl 4-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)piperazine-2-carboxylate.
Step 3: The crude material from above (104 mg, 0.268 mmol) was dissolved in dichloromethane (1.5 mL) and triethylamine (0.080 mL, 0.574 mmol, 2.15 equiv) and propyl chloroformate (0.050 mL, 0.445 mmol, 1.67 equiv) were added. The mixture was stirred for 1 h before quenching by the addition of saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A042 as a yellow solid (88.7 mg, 70%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.6 Hz, 1 H), 7.85 (s, 1 H), 7.58 (d, / = 8.5 Hz, 1 H), 4.86-4.32 (m, 3 H), 4.09-3.85 (m, 3 H), 3.78-3.46 (m, 4 H), 3.16-2.99 (m, 6 H), 1.90 (s, 4 H), 1.64- 1.48 (m, 2 H), 0.94-0.78 (m, 3 H). LCMS (ESI-TOF) nt/z 474.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,5-dimethylpiperazine-l-carboxylate (A043)
Figure imgf000142_0001
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3,5-dimethylpiperazine-l -carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3,5-dimethylpiperazine-l - carboxylate.
Step 2: The resulting intermediate (200 mg, 0.437 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (10 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2,6-dimethylpiperazin- 1 -yl)methanone.
Step 3: The crude material from above (120 mg, 0.335 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 1.73 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.30 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A043 as a white solid (50 mg, 34%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.62 (dd, / = 8.6, 1.5 Hz, 1 H), 4.73-3.62 (m, 6 H), 3.23-2.93 (m, 6 H), 1.90 (s, 4 H), 1.58 (dq, / = 13.9, 7.0 Hz, 2 H), 1.20 (s, 6 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 444.1 [M + H+] with a purity of >95%.
Methyl 9-chloro-6-(4-(propoxycarbonyl)piperazine-l-carbonyl)-l,2,3,4-tetrahydroacridine-3- carboxylate (A044)
Figure imgf000142_0002
Compound A044 was prepared according to General Procedure A, B, and C2 using methyl 3- oxocyclohexanecarboxylate (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.8 Hz, 1 H), 7.97 (s, 1 H), 7.67 (d, / = 10.0 Hz, 1 H), 3.8 (t, / = 6.8 Hz, 2 H), 3.66 (s, 5 H), 3.54-3.31 (m, 6 H), 3.27-3.23 (m, 2 H), 3.09-3.04 (m, 3 H), 2.32 (m, 1 H), 2.01 (br s, 1 H), 1.59-1.57 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) nt/z 474.4 [M + H+] with a purity of >97%.
Propyl 4-(7-(aminomethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A045)
Figure imgf000143_0001
Compound A045 was prepared using A046 as starting material according to General Procedure D.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 1.6 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.66-3.23 (m, 8 H), 3.26-3.00 (m, 4 H), 2.69-2.49 (m, 2 H), 2.09-2.07 (m, 1 H), 1.82-1.46 (m, 6 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 455.4 [M + H+] with a purity of >98%.
4-(9-chloro-7-cyano-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000143_0002
Compound A046 was prepared according to General Procedure A, B and C2 using 4- oxocyclohexanecarbonitrile (General Procedure A) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.22 (d, / = 8.4 Hz, 1 H), 7.98 (s, 1 H), 7.69 (dd, / = 8.4, 1.6 Hz, 1 H), 3.96 (t, / = 6.4 Hz, 2 H), 3.66-3.31 (m, 8 H), 3.21-3.16 (m, 4 H), 2.32-2.16 (m, 2 H), 1.60-1.55 (m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 441.2 [M + H+] with a purity of >98%.
Propyl 4-(6-allyl-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate (A047)
Figure imgf000143_0003
A047 Compound A047 was prepared according to General Procedure A, B, and C2 using 3- allylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.4, 1.6 Hz, 1 H), 5.95-5.85 (m, 1 H), 5.13-5.07 (t, / = 14.4 Hz, 2 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.31 (m, 8 H), 3.17-3.10 (m, 2 H), 2.92-2.83 (m, 1 H), 2.76-2.69 (m, 1 H), 2.19-2.15 (m, 2 H), 2.07-1.95 (m, 2 H), 1.67-1.46 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 456.2 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(methylcarbamoyl)piperazine-l- carboxylate (A048)
Figure imgf000144_0001
Step 1 : To a solution of Compound A042 (300.5 mg, 0.634 mmol) in methanol (2 mL), 1,4-dioxane (1 mL) and water (2 mL) was added lithium hydroxide (34.4 mg, 1.436 mmol, 2.26 equiv). Upon completion, ethyl acetate was added and the pH was adjusted to 3 using concentrated hydrochloric acid. The organic layer was separated and the aqueous phase was extracted with ethyl acetate thrice and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-l- (propoxycarbonyl)piperazine-2-carboxylic acid.
Step 2: To a solution of the crude intermediate (150 mg, 0.326 mmol) in dichloromethane (2 mL) and N,N-dimethylformamide (5 μί) was added oxalyl chloride (0.2 mL, 0.489 mmol, 1.5 equiv). After 2 h, the solvent was removed under reduced pressure. The residue (77.8 mg, 0.163 mmol) was re- dissolved in tetrahydrofuran (1 mL) and triethylamine (32.9 mg, 0.325 mmol, 2 equiv) followed by methylamine (5.05 mg, 0.163 mmol, 1 equiv) were added. After 20 min, saturated ammonium chloride was added and the mixture was extracted with ethyl acetate. The combined organics were washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/dichloromethane) to afford compound A048 as a white solid (2.8 mg, 4%) upon lyophilization.
LCMS (ESI-TOF) m/z 473.2 [M + H+] with a purity of >95%.
4-(9-Chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-N-propylpiperazine-l-carboxamide (A049)
Figure imgf000144_0002
A049 Step 1 : General Procedure CI was performed between commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid and teri-butyl piperazine-l-carboxylate as starting materials to obtain teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate.
Step 2: The resulting intermediate (800 mg, 1.861 mmol) was dissolved in trifluoroacetic acid (2 mL) and dichloromethane (15 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3 -yl) (piper azin- 1 -yl)methanone .
Step 3: Propylamine (11 μL, 0.14 mmol, 1 equiv), N,N-diisoproylethylamine (0.1 mL, 0.57 mmol, 4.07 equiv) and N,N'-disuccinimidyl carbonate (56.3 mg, 0.21 mmol, 1.5 equiv) were dissolved in dichloromethane (1 mL). After 2 h, a solution of intermediate (46.2 mg, 0.14 mmol) and N,N- diisoproylethylamine (0.15 mL, 0.86 mmol, 6.15 equiv) in dichloromethane (2 mL) was added. After 1 h of stirring, the mixture was concentrated and purified by column chromatography (methanol/dichloromethane) to afford A049 as a white solid (30 mg, 52%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.93 (d, / = 1.2 Hz, 1 H), 7.65 (dd, / = 8.6, 1.6 Hz, 1 H), 6.55 (t, / = 5.4 Hz, 1 H), 3.76-3.35 (m, 8 H), 3.11-2.90 (m, 6 H), 1.90 (s, 4 H), 1.51-1.33 (m, 2 H), 0.83 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 415.2 [M + H+] with a purity of >99%. l-(4-(9-Chloro-5,6,7,8-tetrahydroacridine-3-carbon l)piperazin-l-yl)pent-4-en-l-one (A050)
Figure imgf000145_0001
Intermediate (9-chloro-5,6,7,8-tetrahydroacridin-3-yl)(piperazin-l-yl)methanone (0.1 mmol) and NN- diisopropylethylamine (0.175 mL, 1.0 mmol, 10 equiv) were stirred in N,N-dimethylformamide (10 mL) at 4 °C for 15 min before pent-4-enoyl chloride (0.11 mL, 1.0 mmol, 10 equiv) was added dropwise. The reaction was stirred for 2 h at the same temperature before warming to room temperature for another 4 h. The mixture was then quenched with brine and extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative -HPLC to affod A050 (6.55 mg, 16%) as a yellow oil.
*H NMR (400 MHz, DMSO-<¾ δ 8.22 (d, / = 8.6 Hz, 1 H), 7.97 (d, / = 1.2 Hz, 1 H), 7.68 (dd, / = 8.6, 1.5 Hz, 1 H), 5.95-5.77 (m, 1 H), 5.05 (d, / = 17.2 Hz, 1 H), 4.96 (d, / = 9.7 Hz, 1 H), 3.83-3.26 (m, 8 H), 3.08 (s, 2 H), 2.99 (s, 2 H), 2.43 (s, 2 H), 2.25 (dd, / = 13.9, 6.6 Hz, 2 H), 1.91 (d, / = 2.8 Hz, 4 H).
LCMS (ESI-TOF) m/z 412.2 [M + H+] with purity >94%.
3-Methyl 1-propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l,3- dicarboxylate (A051)
Figure imgf000146_0001
Compound A051 was performed according to General Procedure CI between commercially available 9-chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and 3-methyl-l-propylpiperazine-l,3-di carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.21 (d, / = 8.5 Hz, 1 H), 7.90 (s, 1 H), 7.60 (d, / = 8.6 Hz, 1 H), 5.24-4.73 (m, 2 H), 4.41 (d, / = 14.1 Hz, 1 H), 3.97 (t, / = 6.3 Hz, 2 H), 3.90-3.68 (m, 5 H), 3.38-3.20 (m, 3 H), 3.00 (br s, 3 H), 1.91 (s, 4 H), 1.58 (dq, / = 14.3, 7.0 Hz, 2 H), 0.89 (t , / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 474.1 [M + H+] with a purity of >97%.
Propyl 2-(acetoxymethyl)-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A052)
Figure imgf000146_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (1.22 mg, 0.010 mmol, 0.05 equiv) was dissolved in dichloromethane (1 mL) at 0 °C. Triethylamine (83 μL·, 0.60 mmol, 3 equiv) followed by acetic anhydride (22 μL·, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL). The organic layer was separated and washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated to give teri-butyl 2-(acetoxymethyl)-4-(9-chloro-5,6,7,8-tetrahydroacridine- 3-carbonyl)piperazine-l -carboxylate.
Step 3: The resulting crude intermediate (80 mg, 0.159 mmol) was dissolved in trifluoroacetic acid (0.4 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl acetate.
Step 4: The crude material from above (70 mg, 0.174 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 3.34 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 2.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A052 as a white solid (10 mg, 12%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.63 (s, 1 H), 4.59-3.34 (m, 9 H), 3.24-2.74 (m, 6 H), 2.10-1.66 (m, 7 H), 1.67-1.48 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 488.1 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A053)
Figure imgf000147_0001
Compound A053 was prepared according to General Procedure E, F, C2 and G using 3-(pyridin-2- yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.54 (d, / = 4.4 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 7.98 (d, / = 1.2 Hz, 1 H), 7.78 (dt, / = 7.6, 1.6 Hz, 1 H), 7.68 (dd, / = 8.4, 1.6 Hz, 1 H), 7.43 (d, / = 8.0 Hz, 1 H), 7.28-7.25 (m, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.38 (m, 11 H), 3.25-3.05 (m, 2 H), 2.32.-2.27 (m, 1 H), 2.15 (br s, 1 H), 1.61-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 493.5 [M + H+] with a purity of >95%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-((isobutyryloxy)methyl)piperazine- 1-carboxylate (A054)
Figure imgf000147_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (1.22 mg, 0.010 mmol, 0.05 equiv) was dissolved in dichloromethane (1 mL) at 0 °C. Triethylamine (83 μL·, 0.60 mmol, 3 equiv) followed by isobutyryl chloride (25.6 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL). The organic layer was separated and washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3- carbonyl)-2-((isobutyryloxy)methyl)piperazine- 1 -carboxylate. Step 3: The resulting crude intermediate (80 mg, 0.151 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl isobutyrate.
Step 4: The crude material from above (60 mg, 0.14 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 4.16 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.12 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A054 as a white solid (15 mg, 21%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.94 (s, 1 H), 7.63 (s, 1 H), 4.60-3.35 (m, 9 H), 3.24-2.79 (m, 7 H), 1.90 (s, 4 H), 1.69-1.48 (m, 2 H), 1.08 (s, 3 H), 0.96-0.70 (m, 6 H).
LCMS (ESI-TOF) m/z 517.2 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(((3- methoxypropanoyl)oxy)methyl)-piperazine-l-carboxylate (A055)
Figure imgf000148_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (4.9 mg, 0.040 mmol, 0.2 equiv) was dissolved in dichloromethane (5 mL) at 0 °C. Triethylamine (42 μL·, 0.30 mmol, 1.5 equiv) followed by 3-methoxypropionyl chloride (29.4 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL). The organic layer was separated and washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3- carbonyl)-2-(((3-methoxypropanoyl)oxy)methyl)piperazine-l -carboxylate.
Step 3: The resulting crude intermediate (80 mg, 0.147 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl 3-methoxypropanoate. Step 4: The crude material from above (50 mg, 0.112 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 5.19 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.89 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A055 as a white solid (8 mg, 13%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.92 (s, 1 H), 7.63 (s, 1 H), 4.58-3.46 (m, 12 H), 3.23-2.85 (m, 10 H), 1.90 (s, 4 H), 1.66-1.50 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 533.1 [M + H+] with a purity of >94%.
2-carbamoyl-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-
Figure imgf000149_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-carbamoylpiperazine-l-carboxylate to give teri-butyl 2-carbamoyl-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate.
Step 2: The resulting crude intermediate (98.1 mg, 0.207 mmol) was dissolved in trifluoro acetic acid (0.16 mL) and dichloromethane (0.16 mL) for 20 min. The mixture was concentrated under reduced pressure and then diluted with ethyl acetate. The organic layer was washed with saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-2-carboxarnide.
Step 3: The crude material from above (57.8 mg, 0.155 mmol) was dissolved in dichloromethane (1.6 mL) and triethylamine (29 mg, 0.287 mmol, 1.84 equiv) followed by n-propyl chloroformate (24.0 mg, 0.196 mmol, 1.26 equiv) were added at room temperature. The mixture was stirred for 20 min before quenching by the addition of saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (100% ethyl acetate) followed by preparative HPLC to afford A056 as a white solid (24.6 mg, 35%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.17 (d, / = 8.6 Hz, 1 H), 7.87 (s, 1 H), 7.56 (d, / = 8.5 Hz, 1 H), 7.24-6.76 (m, 2 H), 4.49-4.24 (m, 2 H), 3.98 (dd, / = 10.3, 6.3 Hz, 3 H), 3.80 (d, / = 12.8 Hz, 1 H), 3.49 (dd, / = 26.9, 12.2 Hz, 2 H), 3.13 (dd, / = 22.2, 11.8 Hz, 1 H), 3.04 (s, 2 H), 2.99 (s, 2 H), 1.91 (s, 4 H), 1.58 (dq, / = 13.8, 6.8 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.1 [M + H+] with a purity of >97%. Propyl 4-(9-chloro-6-(pyrimidin-5-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A057)
Figure imgf000150_0001
Compound A057 was prepared according to General Procedure A, B and C2 using 3-(pyrimidin-5- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 9.11 (s, 1 H), 8.87 (s, 2 H), 8.24 (d, / = 8.8 Hz, 1 H), 7.98 (d, / = 1.6 Hz, 1 H), 7.69 (dd, / = 8.8, 1.6 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.41-3.33 (m, 10 H), 3.26-3.24 (m, 2 H), 3.10-3.06 (m, 1 H), 2.33-2.32 (m, 1 H), 2.21-2.10 (m, 1 H), 1.61-1.58 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 494.2 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-6-(pyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A058)
Figure imgf000150_0002
Compound A058 was prepared according to General Procedure A, B and C2 using 3-(pyridin-4- yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.54 (d, / = 5.6 Hz, 2 H), 8.23 (d, / = 8.8 Hz, 1 H), 7.98 (s, 1 H), 7.68 (d, / = 1.6 Hz, 1 H), 7.41 (d, / = 5.6 Hz, 2 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.67-3.35 (m, 8 H), 3.24-3.20 (m, 4 H), 3.10-3.01(m,l H), 2.23 (br s, 1 H), 2.07-2.04 (m, 1 H),1.60-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 493.2 [M + H+] with a purity of >97%.
Propyl 3-carbamoyl-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A059)
Figure imgf000150_0003
A059 Compound A059 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl 3-carbamoylpiperazine-l- carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.18 (d, / = 8.5 Hz, 1 H), 7.94 (s, 1 H), 7.62 (d, / = 8.5 Hz, 1 H), 7.45-6.83 (m, 2 H), 5.13-4.22 (m, 2 H), 4.05-3.90 (m, 2 H), 3.81 (br s, 1 H), 3.47 (br s, 1 H), 3.30 (d, / = 9.8 Hz, 1 H), 3.11-2.91 (m, 5 H), 2.52 (s, 1 H), 1.91 (s, 4 H), 1.64-1.50 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.1 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2-hydroxyethyl)piperazine-l- carboxylate (A060)
Figure imgf000151_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(2-hydroxyethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2- hydroxyethyl)piperazine- 1 -carboxylate.
Step 2: The resulting crude intermediate (54.3 mg, 0.115 mmol) was dissolved in trifluoroacetic acid (1.05 mL) and dichloromethane (1.5 mL) for 30 min. The mixture was concentrated under reduced pressure and then diluted with ethyl acetate. The organic layer was washed with saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin-3-yl)(3-(2-hydroxyethyl)piperazin-l-yl)methanone.
Step 3: The crude material from above (16.5 mg, 0.044 mmol) was dissolved in dichloromethane (1 mL) and triethylamine (10.88 mg, 0.108 mmol, 2.44 equiv) followed by n-propyl chloroformate (8.1 mg, 0.066 mmol, 1.5 equiv) were added at room temperature. After 20 min, saturated ammonium chloride was added and the aqueous layer was extracted with dichloromethane. The organic layers were washed with brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A060 as a white solid (4.88 mg, 24%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.92 (s, 1 H), 7.64 (d, / = 6.2 Hz, 1 H), 4.70-3.69 (m, 6 H), 3.17-2.84 (m, 9 H), 1.90 (s, 4 H), 1.83-1.37 (m, 5 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 460.1 [M + H+] with a purity of >99%.
Propyl 2-((butyryloxy)methyl)-4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A061)
Figure imgf000152_0001
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(hydroxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: Intermediate from Step 1 (92 mg, 0.20 mmol) and 4-dimethylaminopyridine (4.9 mg, 0.040 mmol, 0.2 equiv) was dissolved in dichloromethane (5 mL) at 0 °C. Triethylamine (30.4 μL·, 0.30 mmol, 1.5 equiv) followed by butyryl chloride (25.6 mg, 0.24 mmol, 1.2 equiv) was added. The reaction mixture was stirred for 15 min before diluting with dichloromethane (50 mL). The organic layer was separated and washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford teri-butyl 2-((butyryloxy)methyl)-4-(9-chloro- 5,6,7, 8-tetrahydroacridine-3-carbonyl)piperazine-l -carboxylate.
Step 3: The resulting crude intermediate (80 mg, 0.151 mmol) was dissolved in trifluoroacetic acid (0.5 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (4-(9-chloro-5, 6,7,8- tetrahydroacridine-3-carbonyl)piperazin-2-yl)methyl butyrate.
Step 4: The crude material from above (50 mg, 0.116 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 5 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 3.75 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A061 as a white solid (8 mg, 13%) upon lyophilization.
!H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.64 (s, 1 H), 4.65-3.36 (m, 9 H), 3.24-2.91 (m, 7 H), 1.90 (s, 5 H), 1.58 (dq, / = 14.2, 7.1 Hz, 3 H), 0.97-0.54 (m, 7 H).
LCMS (ESI-TOF) m/z 516.2 [M + H+] with a purity of >99%.
Propyl (25)-4-(9-chloro-7-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A062)
Figure imgf000152_0002
From the intermediate of General Procedure B in the synthesis of A034, General Procedure CI was conducted with (S)-n-propyl 2-methylpiperazine- 1-carboxylate to afford compound A062. *H NMR (400 MHz, DMSO-<¾) δ 8.21 (d, / = 8.6 Hz, 1 H), 7.94 (s, 1 H), 7.67 (s, 1 H), 4.68-3.47 (m, 9 H), 3.26-2.86 (m, 6 H), 2.72-2.55 (m, 3 H), 2.36-2.26 (m, 1 H), 2.12-1.97 (m, 1 H), 1.72-1.45 (m, 3 H), 1.27-0.95 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 502.2 [M + H+] with a purity of >99%.
Propyl (2/?)-4-(9-chloro-7-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A063)
Figure imgf000153_0001
From the intermediate of General Procedure B in the synthesis of A034, General Procedure CI was conducted with (R)-n-propyl 2-methylpiperazine- 1-carboxylate to afford compound A063.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.18 (d, / = 8.5 Hz, 1 H), 7.90 (s, 1 H), 7.61 (d, / = 8.6 Hz, 1 H), 4.47 (s, 1 H), 4.15-3.71 (m, 5 H), 3.48 (s, 3 H), 3.30 (s, 1 H), 3.21-3.11 (m, 2 H), 3.05 (s, 2 H), 3.00 (s, 2 H), 2.74-2.51 (m, 2 H), 1.91 (s, 4 H), 1.64-1.48 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 488.1 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-(trifluoromethyl)piperazine-l- carboxylate (A064)
Figure imgf000153_0002
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-trifluoromethylpiperazine-l - carboxylate to afford teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- (trifluoromethyl)piperazine- 1 -carboxylate.
Step 2: The resulting intermediate (20 mg, 0.40 mmol) was dissolved in trifluoroacetic acid (0.2 mL) and dichloromethane (2 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2-(trifluoromethyl)piperazin-l -yl)methanone.
Step 3: The crude material from above (12 mg, 0.030 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 19.3 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 14.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (30 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A064 as a white solid (5 mg, 34%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.23 (d, / = 8.6 Hz, 1 H), 7.97 (s, 1 H), 7.65 (d, / = 8.5 Hz, 1 H), 5.42 (s, 1 H), 4.55-3.32 (m, 7 H), 3.19-3.00 (m, 5 H), 1.90 (s, 4 H), 1.57 (d, / = 6.4 Hz, 2 H), 0.88 (s, 3 H). 19F NMR (376 MHz, DMSO-<¾) δ -68.79- -69.83 (m, 3 F).
LCMS (ESI-TOF) m/z 484.1 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-6-(cyanomethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A065)
Figure imgf000154_0001
Compound A065 was prepared according to General Procedure A, B and C2 using 2-(3- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.0 Hz, 1 H), 7.96 (d, / = 1.6 Hz, 1 H), 7.67 (dd, / = 8.4, 1.6 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.26 (m, 8 H), 3.25-3.15 (m, 2 H), 2.98-2.84 (m, 2 H), 2.82-2.71 (m, 2 H), 2.32-2.28 (m, 1 H), 2.16-2.12 (m, 1 H), 1.67-1.55 (m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 455.4 [M + H+] with a purity of >96%.
Propyl 4-(7-(2-aminoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A066)
Figure imgf000154_0002
Compound A066 was prepared using A078 as starting material according to General Procedure D.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.4, 1.2 Hz, 1 H), 6.72 (br s, 2 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.70-3.20 (m, 8 H), 3.20-3.00 (m, 3 H), 2.71-2.67 (m, 1 H), 2.53-2.49 (m, 1 H), 2.04-2.01 (m, 2 H), 1.60-1.51 (m, 5 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.2 [M + H+] with a purity of >95%.
Propyl (35)-4-(6-(aminomethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate (A067)
Figure imgf000155_0001
Compound A067 was prepared was prepared according to General Procedure A, B, C2 and D using 2- (3-oxocyclohexyl)acetamide (General Procedure A) and (5) -propyl 3-methylpiperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.8 Hz, 1 H), 7.91 (s, 1 H), 7.63 (dd, / = 8.8, 1.2 Hz, 1 H), 4.01-3.78 (m, 5 H), 3.20-2.84 (m, 7 H), 2.76-2.61 (m, 3 H), 2.10-2.07 (m, 1 H), 1.84 (br s, 1 H), 1.60-1.48 (m, 3 H), 1.17 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.3 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-7-((dimethylamino)methyl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate A068)
Figure imgf000155_0002
Compound A068 was prepared according to General Procedure A, B and C2 using 4- ((dimethylamino)methyl)cyclohexanone (General Procedure A) and n-propyl piperazine-1- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.8, 1.6 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.71-3.26 (m, 8 H), 3.25-3.20 (m, 1 H), 3.21-3.06 (m, 2 H), 2.55-2.49 (m, 1 H), 2.32-2.03 (m, 10 H), 1.61-1.50 (m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 473.3 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2-methoxy-2-oxoethyl)piperazine- 1-carboxylate (A069)
Figure imgf000155_0003
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(2-methoxy-2-oxoethyl)piperazine- 1-carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(2-methoxy-2- oxoethyl)piperazine-l-carboxylate.
Step 2: The resulting intermediate (251 mg, 0.50 mmol) was dissolved in trifluoroacetic acid (0.77 mL) and dichloromethane (1.2 mL) for 30 min. The mixture was concentrated and ethyl acetate was added. The organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give methyl 2-(4-(9-chloro- 5,6,7, 8-tetrahydroacridine-3-carbonyl)piperazin-2-yl)acetate.
Step 3: The crude material from above (201 mg, 0.500 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (101.22 mg, 1.00 mmol, 2 equiv) followed by n-propyl chloroformate (91.4 mg, 0.75 mmol, 1.5 equiv) were added at 0 °C. After 30 min, the mixture was quenched with saturated ammonium chloride and the organic layer was separated. The aqueous layer was extracted with dichloromethane and the combined organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A069 as a white solid (208 mg, 85%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.18 (d, / = 8.5 Hz, 1 H), 7.90 (s, 1 H), 7.61 (d, / = 8.6 Hz, 1 H), 4.47 (s, 1 H), 4.15-3.71 (m, 5 H), 3.48 (s, 3 H), 3.30 (s, 1 H), 3.21-3.11 (m, 2 H), 3.05 (s, 2 H), 3.00 (s, 2 H), 2.74-2.51 (m, 2 H), 1.91 (s, 4 H), 1.64-1.48 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 488.1 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-(2-methoxy-2-oxoethyl)piperazine- 1-carboxylate (A070)
Figure imgf000156_0001
Compound A070 was prepared according to General Procedure CI, using commercially available 9- chloro-5,6,7,8-tetrahydroacridine-3-carboxylic acid and n-propyl 3-(2-methoxy-2- oxoethyl)piperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.18 (d, / = 8.6 Hz, 1 H), 7.91 (s, 1 H), 7.60 (d, / = 8.5 Hz, 1 H), 4.69 (br s, 1 H), 4.15-3.69 (m, 5 H), 3.59 (s, 3 H), 3.23 (dd, / = 13.6, 3.6 Hz, 2 H), 3.06 (s, 2 H), 3.00 (s, 3 H), 2.69 (d, / = 7.3 Hz, 2 H), 1.90 (d, / = 3.0 Hz, 4 H), 1.64-1.46 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 488.1 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-(2-(dimethylamino)-2- oxoethyl)piperazine-l-carboxylate (A071) and propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3- carbonyl)-3-(2-(methylamino)-2-oxoethyl)piperazine-l-carboxylate (A072)
Figure imgf000156_0002
Step 1 : To a solution of A070 (153.4 mg, 0.3144 mmol) in methanol (1 mL) and 1,4-dioxane (0.5 mL) was added an aqueous solution of lithium hydroxide (75.3 mg, 3.144 mmol, 10 equiv) in water (1 mL) at 0 °C. The reaction was allowed to stir for 30 min before quenching by the addition of concentrated hydrochloric acid and ethyl acetate to pH 2. The organic layer was separated and the aqueous layer was extracted thrice with ethyl acetate. The combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 2-(l-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)-4-(propoxycarbonyl)piperazin-2-yl)acetic acid.
Step 2: To a solution of crude intermediate (106.1 mg, 0.2239 mmol) in dichloromethane (2.2 mL) and N,N-dimethylformamide (0.004 mL) was added oxalyl chloride (0.04 mL, 0.466 mmol, 2.08 equiv). When bubbling has ceased (10 min), the suspension was sonicated for 30 min and then stirred for another 1.5 h. The contents were concentrated under reduced pressure. To the resulting residue (36.73 mg, 0.075 mmol) in tetrahydrofuran (1 mL) was added simultaneously triethylamine (0.03 mL, 0.215 mmol, 2.9 equiv) and a 2.0 M solution of dimethylamine (0.05 mL, 0.1 mmol, 1.34 equiv) in ethanol. The mixture was stirred for 3 h before quenching with saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate, the combined organics was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A071 as a yellow solid (21.5 mg, 58%) upon lyophilization.
In a separate pot, the acid chloride residue (36.73 mg, 0.075 mmol) in tetrahydrofuran (1 mL) was added simultaneously triethylamine (0.03 mL, 0.215 mmol, 2.9 equiv) and a 2.0 M solution of methylamine (0.05 mL, 0.1 mmol, 1.34 equiv) in tetrahydrofuran. The mixture was stirred for 18 h before quenching with saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate, the combined oganics was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by preparative HPLC to afford A072 as a white solid (2.2 mg, 6%) upon lyophilization.
A071: *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.17 (d, / = 8.6 Hz, 1 H), 7.91 (s, 1 H), 7.60 (d, / = 8.6 Hz, 1 H), 4.68 (br s, 1 H), 4.07-3.85 (m, 4 H), 3.36-3.12 (m, 2 H), 3.05 (s, 6 H), 3.01-2.67 (m, 8 H), 1.90 (d, / = 3.1 Hz, 4 H), 1.64-1.48 (m, 2 H), 0.87 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 501.2 [M + H+] with a purity of >96%.
A072: *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.17 (d, / = 8.6 Hz, 1 H), 7.90 (s, 1 H), 7.59 (d, / = 8.5 Hz, 1 H), 4.63 (s, 1 H), 4.05-3.79 (m, 4 H), 3.52 (s, 1 H), 3.22-2.89 (m, 5 H), 2.55 (d, / = 4.3 Hz, 3 H), 2.44 (d, / = 7.3 Hz, 1 H), 1.91 (s, 4 H), 1.58 (dq, / = 14.2, 7.2 Hz, 2 H), 0.88 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 487.2 [M + H+] with a purity of >94%.
Propyl (25)-4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (A073)
Figure imgf000158_0001
Intermediate from General Procedure F in the synthesis of A053 was subjected to General Procedure CI with (S)-n-propyl 2-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A073.
*H NMR (400 MHz, DMSO-<¾ δ 8.54 (d, / = 4.6 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.96 (s, 1 H), 7.78 (td, / = 7.7, 1.7 Hz, 1 H), 7.68 (s, 1 H), 7.43 (d, / = 7.9 Hz, 1 H), 7.27 (dd, / = 6.5, 4.9 Hz, 1 H), 4.58-4.04 (m, 2 H), 4.03-3.92 (m, 2 H), 3.91-3.35 (m, 6 H), 3.24-2.83 (m, 4 H), 2.30 (d, / = 20.9 Hz, 1 H), 2.11 (s, 1 H), 1.64-1.49 (m, 2 H), 1.32-0.92 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.2 [M + H+] with a purity of >98%.
Propyl (2/?)-4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A074)
Figure imgf000158_0002
Intermediate from General Procedure F in the synthesis of A053 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A074.
*H NMR (400 MHz, DMSO-<¾ δ 8.54 (d, / = 4.0 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.96 (s, 1 H), 7.78 (td, / = 7.7, 1.7 Hz, 1 H), 7.68 (s, 1 H), 7.43 (d, / = 7.8 Hz, 1 H), 7.26 (dd, / = 7.1, 5.2 Hz, 1 H), 4.52-4.03 (m, 2 H), 4.03-3.92 (m, 2 H), 3.91-3.34 (m, 6 H), 3.24-2.85 (m, 4 H), 2.36-2.22 (m, 1 H), 2.10 (s, 1 H), 1.66-1.49 (m, 2 H), 1.32-0.94 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.2 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-7-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate A075)
Figure imgf000158_0003
Compound A075 was prepared according to General Procedure A, B, C2 using 4-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and n-propyl piperazine- 1-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.8, 1.6 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.66-3.39 (m, 8 H), 3.23-3.00 (m, 3 H), 2.59-2.54 (m, 1 H), 2.38-2.34 (m, 2 H), 2.16 (s, 6 H), 2.11-2.04 (m, 1 H), 1.92 (br s, 1 H), 1.61-1.50 (m, 5 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 487.3 [M + H+] with a purity of >98%.
Propyl 4-(6-(2-aminoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A076)
Figure imgf000159_0001
Compound A076 was prepared using A065 as starting material according to General Procedure D.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.4, 1.6 Hz, 1 H), 3.98 (t, / = 8.0 Hz, 2 H), 3.66-3.60 (m, 8 H), 3.30-3.09 (m, 4 H), 2.93-2.84 (m, 1 H), 2.74-2.67 (m, 3 H), 2.02-1.99 (m, 2 H), 1.61-1.45 (m, 5 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 459.3 [M + H+] with a purity of >97%.
Propyl 4-(6-(2-amino-2-oxoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A077)
Figure imgf000159_0002
Compound A077 was prepared using A065 as starting material in dimethylsulfoxide at 0°C and potassium carbonate followed by 30% hydrogen peroxide was added. The reaction mixture was stirred at room temperature for 12 h. The reaction mass was diluted with ethyl acetate and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate and concentrated to afford A077.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.65 (dd, / = 8.0, 1.6 Hz, 1 H), 7.34 (br s, 1 H), 6.83 (s, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.66-3.26 (m, 8 H), 3.20-3.10 (m, 2 H), 2.99-2.86 (m, 1 H), 2.79-2.72 (m, 1 H), 2.32 (br s, 1 H), 2.19-2.17 (m, 2 H), 2.06-2.03( m, 1 H), 1.61-1.55 (m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 473.3 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-7-(cyanomethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A078)
Figure imgf000160_0001
Compound A078 was prepared according to General Procedure A, B and C2 using 2-(4- oxocyclohexyl)acetonitrile (General Procedure A) and n-propyl piperazine-l-carboxylate for Step 3 (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.21 (d, / = 8.8 Hz, 1 H), 7.95 (s, 1 H), 7.67 (dd, / = 8.4, 1.6 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.66-3.27 (m, 8 H) 3.16-3.11 (m, 3 H), 2.80-2.77 (m, 2 H), 2.71-2.64 (m, 1 H), 2.32-2.28 (m, 1 H), 2.11 (d, / = 10.8 Hz, 1 H), 1.70-1.55 (m, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 455.2 [M + H+] with a purity of >99%.
Propyl 4-(7-(2-amino-2-oxoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A079)
Figure imgf000160_0002
To a solution of compound A078 (91 mg, 0.2 mmol) in dimethylsulf oxide (1 mL) was added potassium carbonate (41.4 mg, 0.3 mmol, 1.5 equiv) and 30% hydrogen peroxide (100 μ¾. The reaction was stirred at room temperature for 24 h before quenching with minimal amount of saturated sodium thiosulfate. The aqueous layer was extracted thrice with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography (methanol/dichloromethane) to afford A079 (50 mg, 53%) as a white solid upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.6 Hz, 1 H), 7.94 (s, 1 H), 7.65 (d, / = 8.7 Hz, 1 H), 7.38 (s, 1 H), 6.85 (s, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.84-3.35 (m, 8 H), 3.25-3.00 (m, 3 H), 2.59 (dd, / = 17.6, 9.5 Hz, 1 H), 2.36-2.16 (m, 3 H), 2.03 (d, / = 12.4 Hz, 1 H), 1.58 (dd, / = 14.0, 6.9 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 473.2 [M + H+] with a purity of >98%.
Propyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-6,8-diazabicyclo[3.2.2]nonane-6- carboxylate (A080)
Figure imgf000160_0003
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 6,8-diazabicyclo[3.2.2]nonane-6- carboxylate to give teri-butyl 8-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-6,8- diazabicyclo[3.2.2]nonane-6-carboxylate.
Step 2: The resulting intermediate (300 mg, 0.638 mmol) was dissolved in trifluoroacetic acid (1.2 mL) and dichloromethane (6 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford (6,8- diazabicyclo[3.2.2]nonan-6-yl)(9-chloro-5,6,7,8-tetrahydroacridin-3-yl)methanone.
Step 3: The crude material from above (100 mg, 0.27 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (58.9 mg, 0.582 mmol, 2.15 equiv) followed by n-propyl chloroformate (53.5 mg, 0.436 mmol, 1.61 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A080 as a white solid (30 mg, 24%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.18 (d, / = 8.5 Hz, 1 H), 7.99 and 7.88 (2 x s, 1 H), 7.62 (s, 1 H), 5.04-3.24 (m, 9 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 2.03-1.71 (m, 6 H), 1.66-1.43 (m, 5 H), 0.89 (s, 3 H).
LCMS (ESI-TOF) m/z 456.2 [M + H+] with a purity of >96%.
Propyl (35)-4-(7-(2-aminoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate (A081)
Figure imgf000161_0001
Compound A081 was prepared according to General Procedure A, B, C2 and D using 2-(4- oxocyclohexyl)acetonitrile (General Procedure A) and (S)-n-propyl 3 -methylpiperazine- 1-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.18 (d, / = 8.8 Hz, 1 H), 7.89 (s, 1 H), 7.61 (d, / = 8.8 Hz, 1 H), 4.21-3.75 (m, 5 H), 3.19-2.99 (m, 5 H), 2.94-2.65 (m, 4 H), 2.54-2.43 (m, 1 H), 2.03-1.95 (m, 2 H), 1.58-1.51 (m, 5 H), 1.16 (d, / = 4.8 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 473.6 [M + H+] with a purity of >95%.
Propyl (35)-4-(9-chloro-7-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate (A082)
Figure imgf000162_0001
Compound A082 was prepared according to General procedures steps A, B and C2 using 4-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (S)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.8 Hz, 1 H), 7.91 (s, 1 H), 7.63 (dd, / = 8.8, 1.6 Hz, 1 H), 4.02-3.77 (m, 5 H), 3.26-2.99 (m, 7 H), 2.59-2.54 (m, 1 H), 2.37-2.32 (m, 2 H), 2.15 (s, 6 H), 2.10-2.03 (m, 1 H), 1.91 (s, 1 H), 1.58-1.48 (m, 5 H), 1.16 (d, / = 4.8 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 501.3 [M + H+] with a purity of >98%.
(5)-(9-Chloro-5,6,7,8-tetrahydroacridin-3-yl)(2-methyl-4-(5-methylisoxazole-3- carbonyl)piperazin-l-yl)methanone A083)
Figure imgf000162_0002
Step 1 : 5-methylisoxazole-3-carboxylic acid (20 mg, 0.15 mmol) and teri-butyl (5)-2- methylpiperazine-l-carboxylate (40.1 mg, 0.20 mmol, 1.33 equiv) were dissolved in N,N- dimethylformamide (10 mL) before HATU (190 mg, 0.5 mmol, 3.33 equiv), N,N- diisopropylethylamine (0.35 mL, 2.0 mmol, 13.3 equiv) and 4-dimethylaminopyridine (1 mg) were added. The mixture was stirred for 4 h at room temperature before adding brine and extracting with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give teri-butyl (5)-2-methyl-4-(5-methylisoxazole-3- carbonyl)piperazine-l-carboxylate.
Step 2: The crude material was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (5 mL) for 4 h before concentrating. The crude material was purified by column chromatography (ethyl acetate/hexanes) to give (5)-(5-methylisoxazol-3-yl)(3-methylpiperazin-l-yl)methanone (15 mg, 48% over 2 steps).
Step 3: The intermediate from above was subjected to General Procedure CI with 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid to afford A083.
*H NMR (400 MHz, DMSO-<¾ δ 8.21 (d, / = 8.5 Hz, 1 H), 7.95 (s, 1 H), 7.66 (d, / = 8.4 Hz, 1 H), 6.49 (d, / = 21.4 Hz, 1 H), 4.58-3.76 (m, 7 H), 3.07 (s, 2 H), 2.99 (s, 2 H), 2.46 (s, 3 H), 1.90 (s, 4 H), 1.23-1.08 (m, 3H).
LCMS (ESI-TOF) m/z 453.2 [M + H+] with purity >96%. Propyl (3S)-4-(9-chloro-7-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A084)
Figure imgf000163_0001
From the intermediate of General Procedure B in the synthesis of A034, General Procedure CI was conducted with (S)-n-propyl 3-methylpiperazine- 1-carboxylate to afford compound A084.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.7 Hz, 1 H), 7.89 (s, 1 H), 7.61 (d, / = 8.7 Hz, 1 H), 4.33 (s, 1 H), 4.03-3.74 (m, 5 H), 3.66 (s, 3 H), 3.33-2.89 (m, 8 H), 2.65 (dd, / = 17.3, 10.4 Hz, 1 H), 2.31 (s, 1 H), 2.06 (d, / = 14.6 Hz, 1 H), 1.73-1.51 (m, 3 H), 1.18 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 502.2 [M + H+] with a purity of >98%.
Propyl (3 ?)-4-(9-chloro-7-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A085)
Figure imgf000163_0002
A085
From the intermediate of General Procedure B in the synthesis of A034, General Procedure CI was conducted with (R)-n-propyl 3-methylpiperazine- 1-carboxylate to afford compound A085.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.90 (s, 1 H), 7.61 (d, / = 8.3 Hz, 1 H), 4.37-4.28 (m, 1 H), 4.04-3.76 (m, 6 H), 3.66 (s, 3 H), 3.32-3.09 (m, 5 H), 3.01-2.91 (m, 1 H), 2.65 (dd, / = 17.2, 10.4 Hz, 1 H), 2.31 (s, 1 H), 2.11-2.00 (m, 1 H), 1.70-1.51 (m, 4 H), 1.18 (d, / = 6.8 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 502.1 [M + H+] with a purity of >95%.
Propyl (35)-4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A086)
Figure imgf000163_0003
Intermediate from General Procedure F in the synthesis of A053 was subjected to General Procedure CI with (S)-n-propyl 3-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A086. *H NMR (400 MHz, DMSO-<¾ δ 8.54 (d, / = 4.0 Hz, 1 H), 8.22 (d, / = 8.6 Hz, 1 H), 7.95 (d, / = 1.2 Hz, 1 H), 7.78 (td, / = 7.7, 1.8 Hz, 1 H), 7.65 (dd, / = 8.6, 1.4 Hz, 1 H), 7.43 (d, / = 7.8 Hz, 1 H), 7.29-7.20 (m, 1 H), 4.97-3.34 (m, 9 H), 3.27-2.73 (m, 5 H), 2.35-2.22 (m, 1 H), 2.10 (s, 1 H), 1.58 (dd, / = 14.0, 6.9 Hz, 2 H), 1.17 (d, / = 5.1 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A087)
Figure imgf000164_0001
Intermediate from General Procedure F in the synthesis of A053 was subjected to General Procedure CI with (R)-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A087.
*H NMR (400 MHz, DMSO-<¾) δ 8.54 (d, / = 3.9 Hz, 1 H), 8.22 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.78 (t, / = 6.9 Hz, 1 H), 7.65 (d, / = 9.0 Hz, 1 H), 7.43 (d, / = 7.8 Hz, 1 H), 7.32-7.17 (m, 1 H), 4.78-3.36 (m, 9 H), 3.21-2.86 (m, 5 H), 2.28 (s, 1 H), 2.10 (s, 1 H), 1.65-1.49 (m, 2 H), 1.16 (s, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >98%.
Propyl (35)-4-(6-(2-aminoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A088)
Figure imgf000164_0002
Compound A088 was prepared was prepared according to General procedure steps A, B, C2 and D using 2-(3-oxocyclohexyl)acetamide (General Procedure A) and (S)-n-propyl 3-methylpiperazine-l - carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.20 (d, / = 8.8 Hz, 1 H), 7.91 (s, 1 H), 7.63 (d, / = 8.4 Hz, 1 H), 4.62-4.10 (m, 3 H), 3.99 (t, / = 6.4 Hz, 2 H), 3.78 (br s, 2 H), 3.20-3.11 (m, 5 H), 2.94-2.66 (m, 5 H), 2.21-2.07 (m, 2 H), 1.60-1.52 (m, 5 H), 1.16 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 473.3 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-7-((5-methyl-l,2,4-oxadiazol-3-yl)methyl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate (A089)
Figure imgf000165_0001
Compound A089 was prepared according to General Procedure A, B and C2 using 4-((5-methyl- l,2,4-oxadiazol-3-yl)methyl)cyclohexanone (General Procedure A) and n-propyl piperazine-1- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.65 (d, / = 7.2 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.67-3.26 (m, 8 H), 3.22-3.04 (m, 3 H), 2.94-2.67 (m, 3 H), 2.66 (s, 3 H), 2.32 (br s, 1 H), 2.04 (d, / = 10.8 Hz, 1 H), 1.67-1.55 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 512.26 [M + H+] with a purity of >99%.
Propyl (35)-4-(9-chloro-6-(guanidinomethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A090)
Figure imgf000165_0002
Compound A067 (0.025 mmol) was stirred in N,N-dimethylformamide (2 mL) before 5- methylthioisourea hemisulfate salt (0.2mmol), N,N-diisopropylethylamine (349 μL·, 2.0 mmol) were added and left to react for 2 h at 80 °C. Upon cooling, the reaction mixture was filtered to give a yellow solution, which was concentrated to dryness. The crude material was purified by preparative HPLC to obtain compound A090 as a yellow oil (trifluoroacetate salt) (28%).
LCMS (ESI-TOF) m/z 501.2 [M + H+] with a purity of > 95%.
Propyl (35)-4-(9-chloro-6-(2-guanidinoethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A091)
Figure imgf000165_0003
Compound A088 (0.025 mmol) was stirred in N,N-dimethylformamide (2 mL) before 5- methylthioisourea hemisulfate salt (0.2mmol), N,N-diisopropylethylamine (349 μL·, 2.0 mmol) were added and left to react for 2 h at 80 °C. Upon cooling, the reaction mixture was filtered to give a yellow solution, which was concentrated to dryness. The crude material was purified by preparative HPLC to obtain compound A091 as a yellow oil (trifluoroacetate salt) (39%). ¾ NMR (400 MHz, CD3OD) δ 8.42 (d, / = 8.7 Hz, 1 H), 8.02 (s, 1 H), 7.75 (dd, / = 8.6, 1.2 Hz, 1 H), 4.36-3.79 (m, 5 H), 3.59 - 3.13 (m, 7 H), 3.10-2.83 (m, 3 H), 2.28-2.16 (m, 1 H), 2.15-2.01 (m, 1 H), 1.78 (dd, / = 14.4, 7.1 Hz, 2 H), 1.73-1.59 (m, 3 H), 1.31 (d, / = 5.9 Hz, 3 H), 0.96 (t, / = 7.4 Hz, 3 H). LCMS (ESI-TOF) nt/z 515.3 [M + H+] with a purity of >98%.
Propyl (35)-4-(9-chloro-6-(pyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A092)
Figure imgf000166_0001
Intermediate from General Procedure F in the synthesis of A058 was subjected to General Procedure CI with (S)-n-propyl 3-methylpiperazine-l -carboxylate as reagent, followed by General Procedure G to afford A092.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.52 (d, / = 5.3 Hz, 2 H), 8.22 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.63 (d, / = 8.6 Hz, 1 H), 7.37 (d, / = 5.2 Hz, 2 H), 4.33 (s, 1 H), 4.07-3.70 (m, 5 H), 3.44-2.90 (m, 8 H), 2.26 (s, 1 H), 2.07 (s, 1 H), 1.66-1.44 (m, 2 H), 1.18 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(9-chloro-6-(pyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A093)
Figure imgf000166_0002
Intermediate from General Procedure F in the synthesis of A058 was subjected to General Procedure CI with (R)-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A093.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.52 (d, / = 5.2 Hz, 2 H), 8.22 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.63 (d, / = 8.5 Hz, 1 H), 7.37 (d, / = 5.3 Hz, 2 H), 4.33 (s, 1 H), 4.06-3.72 (m, 5 H), 3.43-2.93 (m, 8 H), 2.25 (s, 1 H), 2.07 (s, 1 H), 1.65-1.50 (m, 2 H), 1.18 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-6-(5-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A094)
Figure imgf000166_0003
A094 Compound A094 was prepared according to General Procedure E, F, C2 and G using 3-(5- fluoropyridin-2-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (d, / = 3.2 Hz, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.97 (s, 1 H), 7.75-7.66 (m, 2 H), 7.54-7.51 (m, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.67-3.29 (m, 11 H), 3.18-3.12 (m, 1 H), 3.09-3.00 (m, 1 H), 2.33-2.26 (m, 1 H), 2.10-2.06 (m, 1 H), 1.60-1.55 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) m/z 511.3 [M + H+] with a purity of >98%.
Propyl (35)-4-(9-chloro-6-(5-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A095)
Figure imgf000167_0001
Intermediate from General Procedure F in the synthesis of A094 was subjected to General Procedure C2 with (S)-n-propyl 3-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A095.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (d, / = 3.2 Hz, 1 H), 8.22 (d, / = 8 Hz, 1 H), 7.95 (s, 1 H), 7.75-7.64 (m, 2 H), 7.54-7.51 (m, 1 H), 3.98-3.79 (m, 5 H), 3.50-3.28 (m, 4 H), 3.18-3.0 (m, 5 H), 2.32-2.26 (m, 1 H), 2.06 (m,l H), 1.57 (d, / = 6.8 Hz, 2 H), 1.16 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.6 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-6-(3-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A096)
Figure imgf000167_0002
Compound A096 was prepared according to General Procedure E, F, C2 and G using 3-(3- methylpyridin-2-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.35 (d, / = 4.4 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 7.97 (s, 1 H), 7.67 (d, / = 8.8 Hz, 1 H), 7.59 (d, / = 6.8 Hz, 1 H), 7.18-7.15 (m, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.67-3.35 (m, 10 H), 3.25-3.07 (m, 4 H), 2.39 (s, 3 H), 2.13-2.07 (m, 2 H), 1.59-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.6 [M + H+] with a purity of >96%. Propyl (25)-4-(9-chloro-6-(5-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A097)
Figure imgf000168_0001
Intermediate from General Procedure F in the synthesis of A094 was subjected to General Procedure CI with (Sj-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A097.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (d, / = 2.9 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.96 (s, 1 H), 7.77-7.62 (m, 2 H), 7.52 (dd, / = 8.7, 4.5 Hz, 1 H), 4.54-3.34 (m, 10 H), 3.25-2.88 (m, 4 H), 2.27 (s, 1 H), 2.09 (s, 1 H), 1.65-1.51 (m, 2 H), 1.29-0.94 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.2 [M + H+] with a purity of >99%.
Propyl (2/?)-4-(9-chloro-6-(5-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (A098)
Figure imgf000168_0002
Intermediate from General Procedure F in the synthesis of A094 was subjected to General Procedure CI with (RJ-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A098.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (d, / = 2.9 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.96 (br s, 1 H), 7.77-7.61 (m, 2 H), 7.52 (dd, / = 8.7, 4.5 Hz, 1 H), 4.53-3.35 (m, 9 H), 3.24-2.86 (m, 5 H), 2.35- 2.20 (m, 1 H), 2.16-1.98 (m, 1 H), 1.65-1.50 (m, 2 H), 1.29-0.95 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.2 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(3-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A099)
Figure imgf000168_0003
Compound A099 was prepared according to General Procedure E, F, C2 and G using 3-(3- fluoropyridin-2-yl) cyclohexanone (General Procedure E) and n-propyl piperazine- 1-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.38 (d, / = 4.4 Hz, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.97 (s, 1 H), 7.74-7.66 (m, 2 H), 7.41-7.37 (m, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.71-3.38 (m, 10 H), 3.19-3.08 (m, 3 H), 2.23-2.12 (m, 2 H), 1.61-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 511.3 [M + H+] with a purity of >97%.
Propyl (35)-4-(9-chloro-6-(3-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A100)
Figure imgf000169_0001
Intermediate from General Procedure F in the synthesis of A099 was subjected to General Procedure C2 with (S)-n-propyl 3-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A100.
*H NMR (400 MHz, DMSO-<¾) δ 8.39 (d, / = 4.4 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 7.94 (s, 1 H), 7.74-7.64 (m, 2 H), 7.41-7.37 (m, 1 H), 3.99-3.71 (m, 7 H), 3.49-3.42 (m, 1 H), 3.16-2.97 (m, 6 H), 2.27-2.26 (m, 1 H), 2.14-2.12 (m, 1 H), 1.60-1.55 (m, 2 H), 1.17 (br s, 3 H) 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.2 [M + H+] with a purity of >99%.
Propyl ( ? 5)-4-(9-chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A053 Ent-1/2))
Figure imgf000169_0002
A053 (Ent-1 or Ent-2)
Compound A053(Ent-l) was isolated from SFC purification of A053.
*H NMR (400 MHz, DMSO-<¾) δ 8.55 (d, / = 4.8 Hz, 1 H), 8.20 (d, / = 8.4 Hz, 1 H), 7.96 (s, 1 H), 7.84-7.69 (m, 1 H) 7.66 (dd, / = 1.6, 8.8 Hz, 1 H), 7.46 (d, / = 7.2 Hz, 1 H), 7.34-7.26 (m, 1 H), 3.96 (t, / = 6.6 Hz, 2 H), 3.70-3.00 (m, 13 H), 2.27 (br s, 1 H), 2.15-2.05 (m, 1 H), 1.65-1.50 (m, 2 H), 0.87 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 493.3 [M + H+] with a purity of >98%.
Compound A053 (Ent-2) was isolated from SFC purification of A053.
*H NMR (400 MHz, DMSO-<¾ δ 8.54 (d, / = 4.0 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 7.98 (s, 1 H), 7.78 (t, / = 7.6 Hz, 1 H) 7.67 (d, / = 8.8 Hz, 1 H), 7.42 (d, / = 7.6 Hz, 1 H), 7.26 (t, / = 6.0 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.75-3.00 (m, 13 H), 2.28 (br s, 1 H), 2.11 (br s, 1 H), 1.65-1.52 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H). LCMS (ESI-TOF) nt/z 493.3 [M + H+] with a purity of >99%.
Propyl (35)-4-(9-chloro-6-(3-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine-l-carboxylate A101)
Figure imgf000170_0001
Intermediate from General Procedure F in the synthesis of A096 was subjected to General Procedure CI with (S)-n-propyl 3-methylpiperazine-l -carboxylate as reagent, followed by General Procedure G to afford A101.
*H NMR (400 MHz, DMSO-<¾) δ 8.36 (d, / = 4.4 Hz, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.66-7.59 (m, 2 H), 7.18-7.15 (m, 1 H), 3.99-3.77 (m, 5 H), 3.57-3.54 (m, 1 H), 3.48-3.35 (m, 1 H), 3.30-2.97 (m, 7 H), 2.39 (s, 3 H), 2.14-2.07 (m, 2 H), 1.60-1.55 (m, 2 H),1.17 (s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 521.3 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(6-(aminomethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A102)
Figure imgf000170_0002
Compound A102 was prepared according to General Procedure A, B, C2 and D using 2-(3- oxocyclohexyl)acetamide (General Procedure A) and (R)-n-propyl 3-methylpiperazine- 1-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.0 Hz, 1 H), 7.91 (s, 1 H), 7.63 (d, / = 8.4 Hz, 1 H), 3.98-3.97 (m, 2 H), 3.79 (br s, 1 H), 3.30-2.87 (m, 7 H), 2.75-2.63 (m, 3 H), 2.49 (m, 2 H), 2.15-2.08 (m, 1 H), 1.92-1.61 (m, 1 H), 1.58-1.56 (m, 3 H), 1.16 (m, 3 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) nt/z 459.31 [M + H+] with a purity of >95%.
Propyl 4-(9-chloro-6-(pyridin-2-ylmethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A103)
Figure imgf000170_0003
Compound A103 was prepared according to General Procedure E, F, C2 and G using 3-(pyridin-2- ylmethyl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (d, / = 4.4 Hz, 1 H), 8.18 (d, / = 8.4 Hz, 1 H), 7.91 (s, 1 H), 7.76-7.72 (m, 1 H), 7.64 (d, / = 8.0 Hz, 1 H), 7.32 (d, / = 8.0 Hz, 1 H), 7.25-7.22 (m, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.65-3.26 (m, 8 H), 3.17-3.12 (m, 1 H), 3.07-3.02 (m, 1 H), 2.93-2.78 (m, 4 H), 2.42-2.40 (m,l H), 2.05-2.02 (m, 1 H), 1.64-1.55 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.3 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(2-methylpyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A104)
Figure imgf000171_0001
Compound A104 was prepared according to the General Procedure E, F, C2 and G using 3-(2- methylpyridin-4-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.39 (d, / = 5.6 Hz, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 7.97 (s, 1 H), 7.68 (d, / = 8.4 Hz, 1 H ), 7.27 (s, 1 H), 7.19 (d, / = 5.2 Hz, 1 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.66-3.26 (m, 7 H), 3.20-2.99 (m, 6 H), 3.47 (s, 3 H), 2.21 (br s, 1 H), 2.07-2.01 (m, 1 H), 1.61-1.55 (m, 2 H), 0.89 (t, / =7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.3 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate A105)
Figure imgf000171_0002
Compound A105 was prepared according to General Procedure A, B and C2 using 3-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (d, / = 8.8 Hz, 1 H), 3.97 (t, / = 6.4 Hz, 2 H), 3.70-3.35 (m, 8 H), 3.20-2.92 (m, 2 H), 2.92-2.83 (m, 1 H), 2.76-2.69 (m, 1 H), 2.37-2.31 (m, 2 H), 2.14 (s, 6 H), 2.07-2.04 (m, 1 H), 1.94-1.93 (m, 1 H) 1.60-1.47 (m, 5 H), 0.95 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 487.2 [M + H+] with a purity of >96%. Propyl (3 ?)-4-(9-chloro-6-(5-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A106)
Figure imgf000172_0001
Intermediate from General Procedure F in the synthesis of A094 was subjected to General Procedure CI with (RJ-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A106.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.48 (d, / = 2.5 Hz, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 7.92 (s, 1 H), 7.71-7.57 (m, 2 H), 7.48 (dd, / = 8.6, 4.4 Hz, 1 H), 4.34 (s, 1 H), 4.09-3.70 (m, 5 H), 3.49-3.29 (m, 3 H), 3.27-3.12 (m, 3 H), 3.12-2.91 (m, 2 H), 2.39-2.19 (m, 1 H), 2.18-2.00 (m, 1 H), 1.71-1.50 (m, 2 H), 1.18 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.2 [M + H+] with a purity of >99%.
Propyl (25)-4-(9-chloro-6-(3-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A107)
Figure imgf000172_0002
Intermediate from General Procedure F in the synthesis of A096 was subjected to General Procedure CI with (SJ-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A107.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.34 (d, / = 4.6 Hz, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 7.93 (s, 1 H), 7.64 (d, / = 8.5 Hz, 1 H), 7.56 (d, / = 7.6 Hz, 1 H), 7.13 (dd, / = 7.4, 4.8 Hz, 1 H), 4.42-3.67 (m, 6 H), 3.65-3.37 (m, 2 H), 3.37-3.07 (m, 6 H), 2.39 (s, 3 H), 2.22-1.98 (m, 2 H), 1.64-1.53 (m, 2 H), 1.11 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl (2/?)-4-(9-chloro-6-(3-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A108)
Figure imgf000172_0003
A108 Intermediate from General Procedure F in the synthesis of A096 was subjected to General Procedure CI with (RJ-n-propyl 2-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A108.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.34 (d, / = 4.6 Hz, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 7.93 (s, 1 H), 7.64 (d, / = 8.5 Hz, 1 H), 7.56 (d, / = 7.6 Hz, 1 H), 7.13 (dd, / = 7.4, 4.8 Hz, 1 H), 4.42-3.67 (m, 6 H), 3.65-3.37 (m, 2 H), 3.37-3.07 (m, 6 H), 2.39 (s, 3 H), 2.22-1.98 (m, 2 H), 1.64-1.53 (m, 2 H), 1.11 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(9-chloro-6-(3-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A109)
Figure imgf000173_0001
Intermediate from General Procedure F in the synthesis of A096 was subjected to General Procedure CI with (RJ-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A109.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.34 (d, / = 4.4 Hz, 1 H), 8.21 (d, / = 8.6 Hz, 1 H), 7.91 (s, 1 H), 7.62 (d, / = 8.6 Hz, 1 H), 7.56 (d, / = 7.4 Hz, 1 H), 7.13 (dd, / = 7.5, 4.8 Hz, 1 H), 4.34 (br s, 1 H), 4.06-3.71 (m, 5 H), 3.58 (dd, / = 12.3, 7.8 Hz, 1 H), 3.46 (dd, / = 17.1, 10.5 Hz, 1 H), 3.20 (dd, / = 24.6, 14.2 Hz, 4 H), 3.13-2.91 (m, 2 H), 2.39 (s, 3 H), 2.23-2.02 (m, 2 H), 1.65-1.53 (m, 2 H), 1.19 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-(methoxymethyl)piperazine-l- carboxylate (A110)
Figure imgf000173_0002
Step 1 : According to General Procedure CI, commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-(methoxymethyl)piperazine-l- carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- (methoxymethyl)piperazine-l-carboxylate.
Step 2: The resulting intermediate (150 mg, 0.316 mmol) was dissolved in trifluoroacetic acid (1.2 mL) and dichloromethane (6 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5, 6,7,8- tetrahydroacridin-3-yl)(3-(methoxymethyl)piperazin-l-yl)methanone.
Step 3: The crude material from above (100 mg, 0.267 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (54.1 mg, 0.535 mmol, 2 equiv) followed by n-propyl chloroformate (49.2 mg, 0.401 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A110 as a white solid (100 mg, 81%) upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.67 (dd, / = 21.4, 7.8 Hz, 1 H), 4.64-3.39 (m, 9 H), 3.22-2.84 (m, 9 H), 1.90 (t, / = 2.9 Hz, 4 H), 1.58 (dq, / = 14.1, 7.0 Hz, 2 H), 0.88 (dd, / = 9.5, 5.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 460.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(6-(2-aminoethyl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate Al 11)
Figure imgf000174_0001
Compound Alll was prepared was prepared according to General procedure steps A, B, C2 and D using 2-(3-oxocyclohexyl)acetamide (General Procedure A) and (R)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.4 Hz, 1 H), 7.91 (s, 1 H), 7.63 (d, / = 8.4 Hz, 1 H), 4.61-3.71 (m, 6 H), 3.17-3.11 (m, 4 H), 2.94-2.85 (m, 2 H), 2.74-2.66 (m, 3 H), 2.06-1.98 (m, 2 H), 1.61-1.48 (m, 5 H), 1.17 (br s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 473.42 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-6-(pyrazin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A112)
Figure imgf000174_0002
Compound A112 was prepared according to General Procedure E, F, C2 and G using 3-(pyrazin-2- yl)cyclohexan-l-one (General Procedure E) and n-propyl piperazine- 1-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.76 (d, / = 0.8 Hz, 1 H), 8.62-8.61 (m, 1 H), 8.55 (d, / = 2.4 Hz, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 7.98 (d, / = 1.2 Hz, 1 H), 7.68 (dd, / = 8.4, 1.6 Hz, 1 H), 3.97 (t, / = 6.4 Hz, 2 H), 3.67-3.03 (m, 13 H), 2.33-2.31 (m, 1 H), 2.17-2.09 (m, 1 H), 1.61-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 494.2 [M + H+] with a purity of >98%.
Propyl (3 ?)-4-(9-chloro-6-(3-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate Al 13)
Figure imgf000175_0001
Intermediate from General Procedure F in the synthesis of A099 was subjected to General Procedure CI with (R)-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A109.
*H NMR (400 MHz, DMSO-<¾) δ 8.39 (d, / = 4.6 Hz, 1 H), 8.22 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.72 (t, / = 9.4 Hz, 1 H), 7.66 (d, / = 8.6 Hz, 1 H), 7.39 (dt, / = 8.5, 4.4 Hz, 1 H), 4.81-3.58 (m, 10 H), 3.22-2.83 (m, 4 H), 2.29-2.18 (m, 1 H), 2.18-2.02 (m, 1 H), 1.66-1.44 (m, 2 H), 1.17 (d, / = 5.0 Hz, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 525.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(9-chloro-6-(2-methylpyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate Al 14)
Figure imgf000175_0002
Intermediate from General Procedure F in the synthesis of A104 was subjected to General Procedure CI with (R)-n-propyl 3 -methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A114.
*H NMR (400 MHz, DMSO-<¾ δ 8.39 (d, / = 5.1 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.66 (d, / = 8.3 Hz, 1 H), 7.28 (s, 1 H), 7.19 (d, / = 5.0 Hz, 1 H), 4.92-3.58 (m, 5 H), 3.36-2.85 (m, 9 H), 2.47 (s, 3 H), 2.29-2.14 (m, 1 H), 2.14-1.93 (m, 1 H), 1.67-1.47 (m, 2 H), 1.17 (d, / = 5.6 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H). LCMS (ESI-TOF) nt/z 522.0 [M + H+] with a purity of >98%.
Propyl (2/?)-4-(9-chloro-6-(3-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (Al 15)
Figure imgf000176_0001
Intermediate from General Procedure F in the synthesis of A099 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine-l-carboxylate as reagent, followed by General Procedure G to afford A115.
*H NMR (400 MHz, DMSO-<¾ δ 8.39 (d, / = 4.2 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.75-7.63 (m, 2 H), 7.39 (dt, / = 8.5, 4.4 Hz, 1 H), 4.60-3.55 (m, 5 H), 3.51-2.82 (m, 9 H), 2.29-2.18 (m, 1 H), 2.17-2.04 (m, 1 H), 1.66-1.49 (m, 2 H), 1.33-0.95 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 525.2 [M + H+] with a purity of >98%.
4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-cyanopiperazine-l-carboxylate
Figure imgf000176_0002
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-cyanopiperazine-l -carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-cyanopiperazine-l -carboxylate.
Step 2: The resulting intermediate (90 mg, 0.198 mmol) was dissolved in trifluoroacetic acid (1 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give l-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)piperazine-2-carbonitrile.
Step 3: The crude material from above (82.4 mg, 0.23 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (64 μL·, 0.46 mmol, 2 equiv) followed by n-propyl chloroformate (40 μL·, 0.35 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A116 as a white solid (44.7 mg, 44%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.23 (d, / = 8.6 Hz, 1 H), 8.01 (s, 1 H), 7.67 (d, / = 8.8 Hz, 1 H), 5.53 (s, 1 H), 4.28 (d, / = 14.0 Hz, 1 H), 4.11-3.85 (m, 4 H), 3.35 (dd, / = 14.1 , 3.6 Hz, 1 H), 3.21 (t, / = 12.7 Hz, 1 H), 3.15-2.97 (m, 5 H), 1.91 (s, 4 H), 1.73-1.42 (m, 2 H), 0.91 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 441.1 [M + H+] with a purity of >98%. Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-cyanopiperazine-l-carboxylate (A117)
Figure imgf000177_0001
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 2-cyanopiperazine-l -carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-cyanopiperazine-l -carboxylate.
Step 2: The resulting intermediate (192.8 mg, 0.424 mmol) was dissolved in trifluoroacetic acid (2 mL) and dichloromethane (5 mL) for 24 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-(9-chloro-5,6,7,8- tetrahydroacridine-3-carbonyl)piperazine-2-carbonitrile.
Step 3: The crude material from above (150 mg, 0.423 mmol) was dissolved in dichloromethane (2.0 mL) and triethylamine (120 μL·, 0.846 mmol, 2 equiv) followed by n-propyl chloroformate (71 μΐ., 0.635 mmol, 1.5 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford A117 as a white solid (54.3 mg, 29%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.22 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.64 (d, / = 8.6 Hz, 1 H), 5.28 (s, 1 H), 4.46-3.81 (m, 5 H), 3.42 (d, / = 13.3 Hz, 1 H), 3.25-2.87 (m, 6 H), 1.91 (s, 4 H), 1.69-1.54 (m, 2 H), 0.91 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 441.1 [M + H+] with a purity of >97%.
4-(9-chloro-5-methyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000177_0002
Step 1 : According to General Procedure I, 2-amino-terephthalic acid was cyclized with 2- methylcyclohexanone (2 equiv) to obtain crude 5-methyl-9-oxo-5,6,7,8,9,10-hexahydroacridine-3- carboxylic acid.
Step 2: The resulting intermediate was reacted with n-propyl piperazine-l -carboxylate via General Procedure CI to obtain propyl 4-(5-methyl-9-oxo-5,6,7,8,9,10-hexahydroacridine-3- carbonyl)piperazine-l -carboxylate. Step 3: The material from above was subjected to General Procedure B to afford A118 as a white solid upon lyophilization.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.6 Hz, 1 H), 7.97 (d, / = 1.2 Hz, 1 H), 7.66 (dd, / = 8.6, 1.6 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.79-3.34 (m, 8 H), 3.18-3.07 (m, 1 H), 3.00 (t, / = 6.5 Hz, 2 H), 2.13-1.91 (m, 2 H), 1.83 (ddd, / = 10.0, 9.6, 4.9 Hz, 1 H), 1.73-1.52 (m, 3 H), 1.43 (d, / = 7.0 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 430.2 [M + H+] with a purity of >99%.
4-(9-chloro-5-phenyl-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate
Figure imgf000178_0001
Compound A119 was synthesized using General Procedure I with 2-amino-terephthalic acid and 2- phenylcyclohexanone, followed by General Procedure CI using n-propyl piperazine-l-carboxylate as reagent.
*H NMR (400 MHz, DMSO-<¾ δ 8.24 (d, / = 8.6 Hz, 1 H), 7.84 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.6, 1.5 Hz, 1 H), 7.26 (t, / = 7.4 Hz, 2 H), 7.17 (t, / = 7.3 Hz, 1 H), 7.01 (d, / = 7.2 Hz, 2 H), 4.52 (t, / = 5.6 Hz, 1 H), 3.96 (t, / = 6.6 Hz, 2 H), 3.80-3.34 (m, 8 H), 3.21-2.96 (m, 2 H), 2.23 (td, / = 12.8, 6.2 Hz, 1 H), 2.04 (td, / = 10.7, 5.3 Hz, 1 H), 1.92-1.77 (m, 2 H), 1.65-1.48 (m, 2 H), 0.87 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 492.2 [M + H+] with a purity of >99%.
Propyl (3 ?)-4-(9-chloro-6-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A120)
Figure imgf000178_0002
Compound A120 was prepared according to General Procedure A, B and C2 using 3-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (R)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.4 Hz, 1 H), 7.91 (s, 1 H), 7.64 (d, / = 9.2 Hz, 1 H), 4.25-3.69 (m, 4 H), 3.26-2.84 (m, 7 H), 2.76-2.69 (m, 1 H), 2.39-2.32 (m, 3 H), 2.14 (s, 6 H), 2.07- 2.04 (m, 1 H), 1.98-1.93 (m, 1 H), 1.60-1.47 (m, 5 H), 1.16-1.15 (m, 3 H), 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 501.6 [M + H+] with a purity of >97%. Propyl 4-(9-chloro-6-(2-(pyrrolidin-l-yl)ethyl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate A121)
Figure imgf000179_0001
Compound A121 was prepared according to General Procedure A, B and C2 using 3-(2- (pyrrolidine)ethyl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.94 (s, 1 H), 7.65 (d, / = 8.0 Hz, 1 H), 3.97 (t, / = 6.4 Hz, 2 H), 3.66-3.36 (m, 8 H), 3.21-3.10 (m, 4 H), 2.92-2.86 (m, 3 H), 2.77-2.67 (m, 3 H), 2.05-1.96 (m, 2 H), 1.70-1.55 (m, 9 H), 0.88 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 513.5 [M + H+] with a purity of >95%.
Propyl (2/?)-4-(9-chloro-6-(2-methylpyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A122)
Figure imgf000179_0002
Intermediate from General Procedure F in the synthesis of A104 was subjected to General Procedure CI with (RJ-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A122.
*H NMR (400 MHz, DMSO-<¾) δ 8.39 (d, / = 5.1 Hz, 1 H), 8.24 (d, / = 8.6 Hz, 1 H), 7.96 (br s, 1 H), 7.69 (br s, 1 H), 7.28 (s, 1 H), 7.19 (d, / = 5.8 Hz, 1 H), 4.58-4.04 (m, 2 H), 4.03-3.91 (m, 2 H), 3.91- 3.34 (m, 4 H), 3.27-2.89 (m, 6 H), 2.47 (s, 3 H), 2.21 (br s, 1 H), 2.12-1.95 (m, 1 H), 1.65-1.51 (m, 2 H), 1.29-0.93 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl (25)-4-(9-chloro-6-(2-methylpyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A123)
Figure imgf000179_0003
Intermediate from General Procedure F in the synthesis of A104 was subjected to General Procedure CI with (SJ-n-propyl 2-methylpiperazine- 1-carboxylate as reagent, followed by General Procedure G to afford A123. *H NMR (400 MHz, DMSO-<¾ δ 8.39 (d, / = 5.1 Hz, 1 H), 8.24 (d, / = 8.6 Hz, 1 H), 7.95 (br s, 1 H), 7.68 (br s, 1 H), 7.28 (s, 1 H), 7.20 (d, / = 4.8 Hz, 1 H), 4.60-4.03 (m, 2 H), 4.05-3.92 (m, 2 H), 3.90- 3.43 (m, 4 H), 3.28-2.87 (m, 6 H), 2.47 (s, 3 H), 2.21 (br s, 1 H), 2.12-1.95 (m, 1 H), 1.65-1.47 (m, 2 H), 1.30-0.94 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 521.2 [M + H+] with a purity of >98%.
Propyl (25)-4-(9-chloro-6-(3-fluoropyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A124)
Figure imgf000180_0001
Intermediate from General Procedure F in the synthesis of A099 was subjected to General Procedure CI with (S)-n-propyl 2-methylpiperazine- 1 -carboxylate as reagent, followed by General Procedure G to afford A124.
*H NMR (400 MHz, DMSO-<¾ δ 8.39 (dd, / = 3.2, 1.4 Hz, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 7.95 (br s, 1 H), 7.78-7.58 (m, 2 H), 7.39 (dt, / = 8.5, 4.4 Hz, 1 H), 4.55-4.06 (m, 2 H), 4.05-3.90 (m, 2 H), 3.90-3.34 (m, 6 H), 3.23-2.88 (m, 4 H), 2.29-2.18 (m, 1 H), 2.11 (ddd, / = 16.4, 13.2, 8.3 Hz, 1 H), 1.64-1.51 (m, 2 H), 1.22-0.98 (m, 3 H), 0.88 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 525.2 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3-(hydroxymethyl)piperazine-l- carboxylate (A125)
Figure imgf000180_0002
Step 1 : According to General Procedure CI , commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid was reacted with teri-butyl 3-(hydroxymethyl)piperazine-l - carboxylate to give teri-butyl 4-(9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- (hydroxymethyl)piperazine- 1 -carboxylate.
Step 2: The resulting intermediate (230 mg, 0.5 mmol) was dissolved in trifluoroacetic acid (2 mL) and dichloromethane (5 mL) for 2 h. The mixture was diluted with dichloromethane (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (9-chloro-5,6,7,8-tetrahydroacridin- 3-yl)(2-(hydroxymethyl)piperazin-l -yl)methanone.
Step 3: The crude material from above (180 mg, 0.5 mmol) was dissolved in dichloromethane (5.0 mL) and triethylamine (0.14 mL, 1.0 mmol, 2 equiv) followed by n-propyl chloroformate (57 μL·, 0.5 mmol, 1 equiv) were added at 0 °C. After 1 h, the mixture was diluted with ethyl acetate (50 mL) and the organic layer was washed with saturated sodium bicarbonate, brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude material was purified by preparative- HPLC to afford A125 as a white solid (60 mg, 27%) upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.16 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.63 (d, / = 8.7 Hz, 1 H), 4.66 (s, 1 H), 4.38-3.69 (m, 6 H), 3.63-3.39 (m, 2 H), 3.20-2.89 (m, 6 H), 1.90 (s, 4 H), 1.63- 1.49 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 446.1 [M + H+] with a purity of >97%.
Propyl (35)-4-(9-chloro-6-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A126)
Figure imgf000181_0001
Compound A126 was prepared according to General Procedure A, B and C2 using 3-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (S)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.19 (d, / = 8.4 Hz, 1 H), 7.90 (s, 1 H), 7.64 (d, / = 8.8 Hz, 1 H), 4.00-3.78 (m, 5 H), 3.26-3.10 (m, 3 H), 2.92-2.86 (m, 3 H), 2.76-2.66 (m, 2 H), 2.35-2.33 (m, 2 H), 2.14 (s, 6 H), 2.10-2.04 (m, 1 H), 1.99-1.93 (m, 1 H), 1.60-1.50 (m, 5 H), 1.21-1.15 (m, 3 H) 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 501.6 [M + H+] with a purity of >98%.
Propyl (2/?)-4-(9-chloro-5-methyl-5,6,7,8-tetrahydroacridine-3-carbonyl)-2-methylpiperazine-l- carboxylate (A127)
Figure imgf000181_0002
Compound A127 was synthesized using General Procedure I with 2-amino-terephthalic acid and 2- methylcyclohexanone, followed by General Procedure CI using (R)-n-propyl 2-methylpiperazine-l- carboxylate as reagent.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.18 (d, / = 8.5 Hz, 1 H), 7.93 (s, 1 H), 7.62 (d, / = 8.1 Hz, 1 H), 4.41-3.63 (m, 6 H), 3.36-2.95 (m, 6 H), 2.14-2.03 (m, 1 H), 2.02-1.90 (m, 1 H), 1.90-1.77 (m, 1 H), 1.71-1.52 (m, 3 H), 1.43 (d, / = 7.0 Hz, 3 H), 1.11 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 444.1 [M + H+] with a purity of >98%. Propyl 4-(9-chloro-6-(thiazol-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A128)
Figure imgf000182_0001
Compound A128 was prepared according to General Procedure A, B and CI using 3 3-(thiazol-2- yl)cyclohexan-l-one (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.22 (d, / = 8.6 Hz, 1 H), 7.99 (d, / = 1.2 Hz, 1 H), 7.75 (d, / = 3.3 Hz, 1 H), 7.68 (dd, / = 8.6, 1.5 Hz, 1 H), 7.66 (d, / = 3.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.84-3.34 (m, 11 H), 3.11 (t, / = 7.6 Hz, 2 H), 2.49-2.40 (m, 1 H), 2.22-2.07 (m, 1 H), 1.64-1.54 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 499.1 [M + H+] with a purity of >98%.
Propyl (2/?)-4-(9-chloro-6-(thiazol-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A129)
Figure imgf000182_0002
Compound A129 was prepared according to General Procedure A, B and CI using 3 3-(thiazol-2- yl)cyclohexan-l-one (General Procedure A) and (R)-n-propyl 2-methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.22 (d, / = 8.6 Hz, 1 H), 7.97 (br s, 1 H), 7.75 (dd, / = 3.3, 1.0 Hz, 1 H), 7.72-7.61 (m, 2 H), 4.54-4.03 (m, 2 H), 4.03-3.89 (m, 2 H), 3.89-3.65 (m, 2H), 3.63-3.26 (m, 4 H), 3.24-2.86 (m, 4 H), 2.48-2.39 (m, 1 H), 2.24-2.08 (m, 1 H), 1.67-1.51 (m, 2 H), 1.29-0.96 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 513.1 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-6-(5-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A130)
Figure imgf000182_0003
Compound A130 was prepared according to General Procedure E, F, CI and G using 3-(5- methylpyridin-2-yl)cyclohexan-l-one (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.20 (d, / = 8.5 Hz, 1 H), 7.95 (s, 1 H), 7.65 (d, / = 8.3 Hz, 1 H), 7.56 (d, / = 7.9 Hz, 1 H), 7.28 (d, / = 8.0 Hz, 1 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.75-3.11 (m, 13 H), 2.28 (s, 4 H), 2.08 (br s, 1 H), 1.59 (dd, / = 14.2, 7.3 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >98%.
Propyl (2/?)-4-(9-chloro-6-(5-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine-l-carboxylate A131)
Figure imgf000183_0001
Compound A131 was prepared according to General Procedure E, F, CI and G using 3-(5- methylpyridin-2-yl)cyclohexan-l -one (General Procedure E) and (R)-n-propyl 2-methylpiperazine-l - carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.21 (d, / = 8.6 Hz, 1 H), 7.93 (s, 1 H), 7.64 (d, / = 8.6 Hz, 1 H), 7.56 (d, / = 7.9 Hz, 1 H), 7.28 (d, / = 7.8 Hz, 1 H), 4.56-3.67 (m, 6 H), 3.45- 3.10 (m, 8 H), 2.28 (s, 4 H), 2.16-2.01 (m, 1 H), 1.73-1.47 (m, 2 H), 1.11 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 521.2 [M + H+] with a purity of >97%.
Propyl 4-(9-chloro-6-(4-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A132)
Figure imgf000183_0002
Compound A132 was prepared according to General Procedure E, F, CI and G using 3-(4- methylpyridin-2-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (d, / = 5.1 Hz, 1 H), 8.20 (d, / = 8.6 Hz, 1 H), 7.95 (s, 1 H), 7.64 (d, / = 8.6 Hz, 1 H), 7.23 (s, 1 H), 7.06 (d, / = 4.5 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.69- 3.26 (m, 11 H), 3.17 (dd, / = 14.1 , 8.4 Hz, 2 H), 2.32 (s, 3 H), 2.27 (br s, 1 H), 2.15-2.02 (m, 1 H), 1.66-1.51 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >99%.
Propyl (2/?)-4-(9-chloro-6-(4-methylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (A133)
Figure imgf000184_0001
Compound A133 was prepared according to General Procedure E, F, CI and G using 3-(4- methylpyridin-2-yl)cyclohexanone (General Procedure E) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (d, / = 5.0 Hz, 1 H), 8.21 (d, / = 8.7 Hz, 1 H), 7.93 (s, 1 H), 7.64 (d, / = 8.3 Hz, 1 H), 7.23 (s, 1 H), 7.06 (d, / = 4.5 Hz, 1 H), 4.39-3.65 (m, 6 H), 3.44-3.06 (m, 8 H), 2.32 (s, 3 H), 2.27 (br s, 1 H), 2.17-2.00 (m, 1 H), 1.64-1.52 (m, 2 H), 1.11 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(pyrimidin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A134)
Figure imgf000184_0002
Compound A134 was prepared according to General Procedure E, F, C2 and G using 3-(pyrimidin-4- yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, CDC13) δ 9.19 (s, 1 H), 8.68 (d, / = 5.2 Hz, 1 H), 8.27 (d, / = 8.8 Hz, 1 H), 8.00 (s, 1 H), 7.62 (dd, / = 1.6, 8.4 Hz, 1 H), 7.28 (d, / = 6.4 Hz, 1 H), 4.08 (t, / = 6.4Hz, 2 H), 3.81-3.36 (m, 10 H), 3.34-3.27 (m, 2 H), 3.12-3.03 (m, 1 H), 2.44-2.41 (m, 1 H), 2.22-2.16 (m, 1 H), 1.69- 1.64 (m, 2 H), 0.95 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 494.3 [M + H+] with a purity of >99%.
Propyl (2/?)-4-(9-chloro-6-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (A135)
Figure imgf000184_0003
Compound A135 was prepared according to General Procedure A, B and C2 using 3-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.95-7.88 (m, 1 H), 7.71-7.61 (m, 1 H), 4.50-3.90 (m, 4 H), 3.89-3.39 (m, 2 H), 3.21-2.85 (m, 6 H), 2.76-2.72 (m, 1 H), 2.37-2.31 (m, 2 H), 2.19 (s, 6 H), 2.21-1.93 (m, 2 H), 1.60-1.49 (m, 5 H), 1.17-1.00 (m, 3 H), 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 501.4 [M + H+] with a purity of >97%.
Propyl (25)-4-(9-chloro-6-(2-(dimethylamino)ethyl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A136)
Figure imgf000185_0001
Compound A136 was prepared according to General Procedure A, B and C2 using 3-(2- (dimethylamino)ethyl)cyclohexanone (General Procedure A) and (S)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8Hz, 1 H), 7.92 (br s, 1 H), 7.71-7.60 (m, 1 H), 4.50-3.87 (m, 4 H), 3.72-3.35 (m, 2 H), 3.25-2.84 (m, 6 H), 2.76-2.67 (m, 1 H), 2.39-2.28 (m, 2 H), 2.19 (s, 6 H), 2.20-1.93 (m, 2 H), 1.62-1.48 (m, 5 H), 1.17-1.00 (m, 3 H), 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 501.4 [M + H+] with a purity of >98%.
Propyl (35)-4-(9-chloro-6-(prop-2-yn-l-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A137)
Figure imgf000185_0002
Compound A137 was prepared according to General Procedure A, B and C2 using 3-(prop-2- ynyl)cyclohexanone (General Procedure A) and (5)-n-propyl 3 -methylpiperazine- 1-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.8 Hz, 1 H), 7.92 (s, 1 H), 7.64 (d, / =7.2 Hz,l H), 4.61-3.65 (m, 7 H), 3.24-3.14 (m, 4 H), 2.95-2.80 (m, 4 H), 2.36-2.32 (m, 1 H), 2.22-2.11 (m, 2 H), 1.63-1.55 (m, 3 H), 1.16 (s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.3 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(2-cyanopyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l- carboxylate (A138)
Figure imgf000186_0001
Compound A138 was prepared according to General Procedure E, F, C2 and G using 4-(3- oxocyclohexyl)picolinonitrile (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.72 (d, / = 4.8 Hz, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 8.13 (s, 2 H), 7.98 (s, 1 H), 7.77 (dd, / = 4.8 Hz, 1.6 Hz, 1 H), 7.69 (d, / = 8.4 Hz, 1 H), 3.98 (t, /= 6.4 Hz, 2 H), 3.70-3.31 (m, 10 H), 3.24-3.21 (m, 2 H), 3.09-3.04 (m, 1 H), 2.31-2.21 (m, 1 H), 2.11-2.08 (m, 1 H), 1.60-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 518.3 [M + H+] with a purity of >97%.
Propyl 4-(6-(2-carbamoylpyridin-4-yl)-9-chloro-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate (A139)
Figure imgf000186_0002
Compound A138 was dissolved in dimethylsulfoxide and was cooled to 0 °C. Potassium carbonate (5 equiv) followed by 30% hydrogen peroxide (2 equiv) was added and the reaction mixture was stirred at room temperature for 1 h. The reaction was diluted with ethyl acetate and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford compound A139.
*H NMR (400 MHz, DMSO-<¾) δ 8.60 (d, / = 4.8 Hz, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 8.11 (s, 1 H),
8.06 (s, 1 H), 7.98 (s, 1 H), 7.69-7.61 (m, 3 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.70-3.31 (m, 10 H), 3.25-
3.07 (m, 3 H), 2.31-2.25 (m, 1 H), 2.41-1.91 (m, 1 H), 1.60-1.55 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 536.4 [M + H+] with a purity of >97%.
Propyl (2/?)-4-(9-chloro-6-(2-cyanopyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate (A140)
Figure imgf000186_0003
Compound A140 was prepared according to General Procedure E, F, C2 and G using 4-(3- oxocyclohexyl)picolinonitrile (General Procedure E) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.72 (d, / = 5.2 Hz, 1 H), 8.24 (d, / = 8.8 Hz, 1 H), 8.13 (s, 1 H), 7.96 (br s, 1 H), 7.77 (d, / = 4.4 Hz, 1 H), 7.69 (br s, 1 H), 4.51-3.41 (m, 9 H), 3.25-3.01 (m, 5 H), 2.31-2.21 (m, 1 H), 2.19-2.01 (m, 1 H), 1.62-1.53 (m, 2 H), 1.23-1.00 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 532.5 [M + H+] with a purity of >95%.
Propyl (2/?)-4-(6-(2-carbamoylpyridin-4-yl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine-l-carboxylate A141)
Figure imgf000187_0001
Compound A140 was dissolved in dimethylsulfoxide and was cooled to 0 °C. Potassium carbonate (5 equiv) followed by 30% hydrogen peroxide (2 equiv) was added and the reaction mixture was stirred at room temperature for 1 h. The reaction was diluted with ethyl acetate and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford compound A141.
*H NMR (400 MHz, DMSO-<¾) δ 8.61 (d, / = 4.8 Hz, 1 H), 8.27 (d, / = 9.2 Hz, 1 H), 8.06 (s, 1 H), 7.96 (s, 1 H), 7.91-7.72 (s, 2 H), 7.69-7.63 (m, 2 H), 4.49-3.77 (m, 5 H), 3.51-3.08 (m, 9 H), 2.40- 2.26 (m, 1 H), 2.22-2.02 (m, 1 H), 1.61-1.55 (m, 2 H), 1.25-0.90 (m, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 550.3 [M + H+] with a purity of >96%.
Propyl (2/?)-4-(9-chloro-6-(prop-2-yn-l-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylate A142)
Figure imgf000187_0002
Compound A142 was prepared according to General Procedure A, B and C2 using 3-(prop-2- ynyl)cyclohexanone (General Procedure A) and (R)-n-propyl 2-methylpiperazine- 1-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.8 Hz, 1 H), 7.93 (br s, 1 H), 7.66 (br s, 1 H), 4.51- 3.45 (m, 6 H), 3.25-2.79 (m, 8 H), 2.36-2.34 (m, 2 H), 2.22-2.12 (m, 2 H), 1.63-1.53 (m, 3 H), 1.31- 1.16 (br s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 468.2 [M + H+] with a purity of >98%. Propyl (35)-4-(9-chloro-6-(2-cyanopyridin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate A143)
Figure imgf000188_0001
Compound A143 was prepared according to General Procedure E, F, C2 and G using 4-(3- oxocyclohexyl)picolinonitrile (General Procedure E) and (S)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.73 (d, / = 5.6 Hz, 1 H), 8.24 (d, / = 8.8 Hz, 1 H), 8.14 (s, 1 H), 7.95 (s, 1 H), 7.77 (d, / = 4.0 Hz, 1 H), 7.67 (d, / = 9.6 Hz, 1 H), 4.01-3.71 (m, 3 H), 3.41-3.27 (m, 4 H), 3.21-2.91 (m, 7 H), 2.32-2.20 (m, 1 H), 2.15- 2.09 (m, 1 H),1.60-1.55 (m, 2 H), 1.23-1.10 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 532.2 [M + H+] with a purity of >98%.
Propyl 4-(6-(2-(aminomethyl)pyridin-4-yl)-9-chloro-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carboxylate (A144)
Figure imgf000188_0002
Compound A144 was synthesized according to General Procedure D using compound A138 as starting material.
*H NMR (400 MHz, DMSO-<¾) δ 8.57 (d, / = 5.6 Hz, 1 H), 8.26 (br s, 2 H), 8.21 (d, / = 8.8 Hz, 1 H), 7.95 (s, 1 H), 7.67 (dd, / = 8.8 Hz, 1.6 Hz, 1 H), 7.51 (s, 1 H), 7.42 (d, / = 4.0 Hz, 1 H), 4.18 (t, / = 5.6 Hz, 2 H), 3.96 (t, / = 6.4 Hz, 2 H), 3.70-3.31 (m, 6 H), 3.29-3.09 (m, 5 H), 3.09-3.06 (m, 2 H), 2.25-2.21 (m, 1 H), 2.10-2.04 (m, 1 H), 1.58-1.53 (m, 2 H), 0.89 (t, /= 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 522.3 [M + H+] with a purity of >95%.
Propyl (35)-4-(6-(2-carbamoylpyridin-4-yl)-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylate (A145)
Figure imgf000188_0003
Compound A143 was dissolved in dimethylsulfoxide and was cooled to 0 °C. Potassium carbonate (5 equiv) followed by 30% hydrogen peroxide (2 equiv) was added and the reaction mixture was stirred at room temperature for 1 h. The reaction was diluted with ethyl acetate and the organic layer was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford compound A145.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (d, / = 4.8 Hz, 1 H), 8.23 (d, / = 8.8 Hz, 1 H), 8.11 (s, 1 H), 8.06 (s, 1 H), 7.95 (s, 1 H), 7.68-7.61 (m, 3 H), 4.41-3.71 (m, 5 H), 3.41-3.30 (m, 3 H), 3.25-3.02 (m, 6 H), 2.32-2.26 (m, 1 H), 2.22-2.12 (m, 1 H), 1.60-1.55 (m, 2 H), 1.18-1.10 (m, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 550.2 [M + H+] with a purity of >99%.
Propyl 4-(5-allyl-9-chloro-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine-l-carboxylate (A146)
Figure imgf000189_0001
Compound A146 was synthesized via General Procedure A, B and CI using 2-allylcyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure CI) as reagents.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.6 Hz, 1 H), 7.97 (d, / = 11.4 Hz, 1 H), 7.67 (d, / = 8.5 Hz, 1 H), 5.88 (td, / = 16.9, 7.9 Hz, 1 H), 5.08 (dd, / = 22.3, 13.6 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.55 (d, / = 99.3 Hz, 8 H), 2.97 (dd, / = 63.8, 32.0 Hz, 4 H), 2.48-2.40 (m, 1 H), 1.98 (s, 2 H), 1.84-1.62 (m, 2 H), 1.58 (dd, / = 13.9, 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 456.2 [M + H+] with a purity of >96%.
Propyl 4-(9-chloro-6-(5-ethynylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A147)
Figure imgf000189_0002
Compound A147 was prepared according to General Procedure E, F, C2 and G using 3-(5- ethynylpyridin-2-yl)cyclohexanone (General Procedure E) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.64 (d, / = 1.6 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 7.97 (s, 1 H), 7.89 (dd, / = 2.0, 8.4 Hz, 1 H), 7.67 (d, / = 8.8 Hz, 1 H), 7.47 (d, / = 7.6 Hz, 1 H), 4.39 (s, 1 H), 3.97 (t, / = 6.8 Hz, 2 H), 3.75-2.90 (m, 13 H), 2.30 (br s, 1 H), 2.10 (br s, 1 H), 1.65-1.50 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H)
LCMS (ESI-TOF) m/z 517.3 [M + H+] with a purity of >98%.
Propyl 4-(9-chloro-6-(2-cyanopyrimidin-4-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)piperazine- 1-carboxylate (A148)
Figure imgf000190_0001
Compound A148 was prepared according to General Procedure A, B and C2 using 4-(3- oxocyclohexyl)pyrimidine-2-carbonitrile (General Procedure A) and n-propyl piperazine-1- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.98 (d, / = 5.2 Hz, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.98-7.94 (m, 2 H), 7.68 (dd, / = 1.2, 8.8 Hz, 1 H), 3.99-3.95 (m, 2 H), 3.80-3.40 (m, 10 H), 3.30-2.90 (m, 3 H), 2.40-1.90 (m, 2 H), 1.58 (dd, / = 6.8, 14.4 Hz, 2 H), 0.897 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 519.3 [M + H+] with a purity of >96%.
Propyl (2/?)-4-(9-chloro-6-(5-ethynylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-2- methylpiperazine- 1-carboxylat
Figure imgf000190_0002
Compound A149 was prepared according to General Procedure E, F, C2 and G using 3-(5- ethynylpyridin-2-yl)cyclohexanone (General Procedure E) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.64 (d, / = 1.6 Hz, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.95 (br s, 1 H), 7.90 (dd, / = 2.4, 8.0 Hz, 1 H), 7.68 (br s, 1 H), 7.47 (d, / = 7.6 Hz, 1 H), 4.39 (s, 1 H), 4.40-2.90 (m, 14 H), 2.27 (br s, 1 H), 2.15-2.05 (m, 1 H), 1.65-1.50 (m, 2 H), 1.40-1.10 (m, 3 H), 0.88 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 531.6 [M + H+] with a purity of >98%.
Propyl (35)-4-(9-chloro-6-(5-ethynylpyridin-2-yl)-5,6,7,8-tetrahydroacridine-3-carbonyl)-3- methylpiperazine- 1-carboxylat
Figure imgf000190_0003
Compound A150 was prepared according to General Procedure E, F, C2 and G using 3-(5- ethynylpyridin-2-yl)cyclohexanone (General Procedure E) and (5)-n -propyl 3-methylpiperazine-l- carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.64 (s, 1 H), 8.22 (d, / = 8.8 Hz, 1 H), 7.95 (s, 1 H), 7.89 (dd, / = 2.0 , 8.4 Hz, 1 H), 7.65 (d, / = 8.8 Hz, 1 H), 7.47 (d, / = 7.6 Hz, 1 H), 4.39 (s, 1 H), 4.10-3.60 (m, 5 H), 3.50-2.90 (m, 9 H), 2.27 (br s, 1 H), 2.09 (br s, 1 H), 1.65-1.50 (m, 2 H), 1.17 (br s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H). LCMS (ESI-TOF) nt/z 531.5 [M + H+] with a purity of >99%.
Propyl 4-(9-chloro-6-(l-methyl-lH-pyrazol-4-yl)-5,6,7,8-tetrahydroacridine-3- carbonyl)piperazine-l-carbo
Figure imgf000191_0001
Compound A148 was prepared according to General Procedure A, B and C2 using 3 -(1 -methyl- 1H- pyrazol-4-yl)cyclohexanone (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.20 (d, / = 8.4 Hz, 1 H), 7.97 (d, / = 1.6 Hz, 1 H), 7.68 (dd, / = 1.6, 8.4 Hz, 1 H), 7.60 (s, 1 H), 7.39 (s, 1 H), 3.97 (t, / = 6.4 Hz, 2 H), 3.78 (s, 3 H), 3.70-3.36 (m, 9 H), 3.29-3.01 (m, 4 H), 2.30-2.24 (m, 1 H), 1.90-1.70 (m, 1 H), 1.61-1.55 (m, 2 H), 0.88 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.3 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(pyridin-3-y azine-l-carboxylate (B001)
Figure imgf000191_0002
Compound B001 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3- (pyridin-3-yl)propanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 9.49 (d, / = 2.0 Hz, 1 H), 8.74-8.73 (m, 1 H) 8.69-8.66 (m, 1 H), 8.59 (s, 1 H), 8.32-8.30 (d, / = 8.4 Hz, 1 H), 8.18 (d, / = 1.2 Hz, 1 H), 7.78 (dd, / = 1.6, 8.8 Hz, 1 H), 7.63-7.59 (m, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.70 (br s, 2 H), 3.51-3.36 (m, 6 H), 1.61-1.56 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 439.4 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-phenylquinolin -7-carbonyl)piperazine-l-carboxylate (B002)
Figure imgf000191_0003
Compound B002 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3- phenylpropanoate (General Procedure H) and n-propylpiperazine-l -carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.49 (s, 1 H), 8.34-8.27 (m, 3 H), 8.15 (d, / = 0.8 Hz, 1 H), 7.75 (dd, / = 1.6, 8.8 Hz, 1 H), 7.57 (dd, / = 5.6, 13.2 Hz, 3 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.70-3.37 (m, 8 H), 1.59-1.57 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 438.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-3-methyl-2-phen lquinoline-7-carbonyl)piperazine-l-carboxylate (B003)
Figure imgf000192_0001
Compound B003 was prepared according to General Procedure I, K and CI using propiophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.32 (d, / = 8.8 Hz, 1 H), 8.11 (s, 1 H), 7.68 (dd, / = 8.8, 1.6 Hz, 1 H), 7.57-7.47 (m, 5 H), 4.06 (t, / = 6.4 Hz, 2 H), 3.81-3.45 (m, 8 H), 2.55 (s, 3 H), 1.69-1.63 (m, 2 H), 0.95 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-3-ethyl-2-phe razine-l-carboxylate (B004)
Figure imgf000192_0002
Compound B004 was prepared similar to B003 using the 1-phenylbutan-l-one and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
¾ NMR (400 MHz, CDC13) δ 8.34 (d, / = 8.4 Hz, 1 H), 8.10 (d, / = 1.2 Hz, 1 H), 7.68 (dd, / = 1.6, 8.4 Hz, 1 H), 7.50-7.40 (m, 5 H), 4.06 (t, / = 6.4 Hz, 2 H), 3.91-3.30 (m, 8 H), 2.98-2.93 (m, 2 H), 1.69-1.52 (m, 2 H), 1.17-1.13 (m, 3 H), 0.95 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.3 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(5-methylpyridin-3-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B005)
Figure imgf000192_0003
Compound B005 was prepared according to General Procedure H, K, J and C2 using ethyl 4-(5- methylpyridin-3-yl)-3-oxobutanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾) δ 9.29 (d, / = 2.0 Hz, 1 H), 8.57 (s, 2 H), 8.51 (s, 1 H), 8.30 (d, / = 8.8 Hz, 1 H), 8.18 (d, / = 0.8 Hz, 1 H), 7.78 (dd, / = 1.2, 8.8 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.72- 3.32 (m, 8 H), 2.44 (s, 3 H), 1.61-1.56 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 453.2 [M + H+] with a purity of >97%.
Propyl (5)-4-(4-chloro-3-methyl-2-phenylquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B006)
Figure imgf000193_0001
Intermediate from General Procedure K in the synthesis of B003 was subjected to General Procedure CI with (S)-n-propyl 2-methylpiperazine-l -carboxylate as reagent to afford B006.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.28 (d, / = 8.6 Hz, 1 H), 8.02 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.60 (d, / = 6.8 Hz, 2 H), 7.56-7.47 (m, 3 H), 4.23 (s, 1 H), 3.98 (pd, / = 10.5, 6.6 Hz, 3 H), 3.81 (d, / = 11.7 Hz, 1 H), 3.32-3.07 (m, 4 H), 2.50 (d, / = 4.5 Hz, 3 H), 1.65-1.50 (m, 2 H), 1.12 (d, / = 6.5 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.2 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-3-methyl-2-phenylquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B007)
Figure imgf000193_0002
Intermediate from General Procedure K in the synthesis of B003 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine-l -carboxylate as reagent to afford B007.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.28 (d, / = 8.6 Hz, 1 H), 8.02 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.60 (d, / = 7.6 Hz, 2 H), 7.56-7.46 (m, 3 H), 4.23 (s, 1 H), 3.99 (pd, / = 10.5, 6.4 Hz, 3 H), 3.81 (d, / = 11.1 Hz, 1 H), 3.36-3.07 (m, 4 H), 2.50 (d, / = 5.8 Hz, 3 H), 1.68-1.51 (m, 2 H), 1.12 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.2 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(pyridin-2-yl) zine-l-carboxylate (B008)
Figure imgf000193_0003
Compound B008 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3- (pyridin-2-yl)propanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.79 (d, / = 4.4 Hz, 1 H), 8.72 (s, 1 H), 8.60 (d, / = 8.4 Hz, 1 H), 8.32 (d, / = 8.4 Hz, 1 H), 8.20 (m, 1 H), 8.09-8.04 (m, 1 H), 7.80 (dd, / = 1.6, 8.8 Hz, 1 H), 7.60-7.57 (m, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.76-3.35 (m, 8 H), 1.61-1.56 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 439.5 [M + H+] with a purity of >98%.
Propyl 4-(2-benzyl-4-chloroquino carboxylate (B009)
Figure imgf000194_0001
Compound B009 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-5- phenylpentanoate (General Procedure H) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.21 (d, / = 8.0 Hz, 1 H), 8.04 (m, 1 H), 7.76 (s, 1 H), 7.71 (dd, / = 7.2, 1.6 Hz, 1 H), 7.37-7.29 (m, 4 H), 7.24-7.20 (m, 1 H), 4.29 (s, 2 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.82-3.32 (m, 8 H), 1.61-1.55 (m, 2 H), 0.88 (t, / = 7.0 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.2 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(p-tolyl) -carboxylate (B010)
Figure imgf000194_0002
Compound B010 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3-p- tolylpropanoate (General Procedure H) and n-propyl piperazine-l -carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.45 (s, 1 H), 8.27-8.23 (m, 3 H), 8.12 (d, / = 0.8 Hz, 1 H), 7.73 (dd, / = 1.6, 8.8 Hz, 1 H), 7.38 (d, / = 8.0 Hz, 2 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.69-3.35(m, 8 H), 2.40 (s, 3 H), 1.61-1.56 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(m-tolyl) -carboxylate (B011)
Figure imgf000194_0003
Compound B011 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3- m-tolylpropanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials. *H NMR (400 MHz, DMSO-<¾) δ 8.46 (s, 1 H), 8.27 (d, / = 8.4 Hz, 1 H), 8.16-8.10 (m, 3 H), 7.74 (dd, / = 1.2, 8.4 Hz, 1 H), 7.46 (t, / = 7.6 Hz, 1 H), 7.36 (d, / = 7.2 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.70-3.36 (m, 8 H), 2.44 (s, 3 H), 1.59 (dd, / = 6.8, 13.6 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(o-tolyl)quino -l-carboxylate (B012)
Figure imgf000195_0001
Compound B012 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-4-o- tolylbutanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.31 (d, / = 8.4 Hz, 1 H), 8.12 (s, 1 H), 8.03 (s, 1 H), 7.79 (d, / = 8.4 Hz, 1 H), 7.55 (d, / = 7.2 Hz, 1 H), 7.45-7.30 (m, 3 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.75-3.35 (m, 8 H), 2.41 (s, 3 H), 1.65-1.50 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(pyridin-4-y azine-l-carboxylate (B013)
Figure imgf000195_0002
Compound B013 was prepared according to General Procedure H, K, J and C2 using ethyl 3-oxo-3- (pyridin-4-yl)propanoate (General Procedure H) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.80 (d, / = 6.0 Hz, 2 H), 8.61 (s, 1 H), 8.33 (d, / = 8.8 Hz, 1 H), 8.28 (dd, / = 1.6 , 4.4 Hz, 2 H), 8.20 (s, 1 H), 7.82 (dd, / = 1.6, 8.8 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.81-3.34 (m, 8 H), 1.61-1.56 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 439.2 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-phenylqui lpiperazine-l-carboxylate (B014)
Figure imgf000195_0003
Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with (S)-n-propyl 3-methylpiperazine-l -carboxylate as reagent to afford B014. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37 (s, 1 H), 8.33-8.23 (m, 3 H), 8.10 (s, 1 H), 7.70 (d, / = 8.3 Hz, 1 H), 7.62-7.49 (m, 3 H), 4.36 (br s, 1 H), 4.12-3.73 (m, 6 H), 3.28-3.16 (m, 2 H), 1.60 (dd, / = 14.2, 7.0 Hz, 2 H), 1.21 (d, / = 6.8 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.1 [M + H+] with a purity of >98%.
Propyl ( ?)-4-(4-chloro-2-phenyl lpiperazine-l-carboxylate (B015)
Figure imgf000196_0001
Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with (R)-n-propyl 3-methylpiperazine-l -carboxylate as reagent to afford B015.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37 (s, 1 H), 8.32-8.24 (m, 3 H), 8.10 (s, 1 H), 7.70 (d, / = 8.6 Hz, 1 H), 7.55 (p, / = 6.1 Hz, 3 H), 4.37 (br s, 1 H), 4.08-3.72 (m, 6 H), 3.34-3.13 (m, 2 H), 1.65- 1.53 (m, 2 H), 1.21 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.1 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-3-methyl-2-phenylquinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B016)
Figure imgf000196_0002
Intermediate from General Procedure K in the synthesis of B003 was subjected to General Procedure CI with (S)-n-propyl 3-methylpiperazine-l -carboxylate as reagent to afford B016.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37 (s, 1 H), 8.33-8.23 (m, 2 H), 8.10 (s, 1 H), 7.70 (d, / = 8.3 Hz, 1 H), 7.62 - 7.49 (m, 3 H), 4.36 (br s, 1 H), 4.12-3.73 (m, 6 H), 3.28-3.16 (m, 2 H), 2.51 (s, 3 H), 1.60 (dd, / = 14.2, 7.0 Hz, 2 H), 1.21 (d, / = 6.8 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.2 [M + H+] with a purity of >95%.
Propyl (/?)-4-(4-chloro-3-methyl-2-phenylquinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B017)
Figure imgf000196_0003
Intermediate from General Procedure K in the synthesis of B003 was subjected to General Procedure CI with (R)-n-propyl 3-methylpiperazine-l -carboxylate as reagent to afford B017.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.28 (d, / = 8.6 Hz, 1 H), 8.01 (s, 1 H), 7.70 (d, / = 8.7 Hz, 1 H), 7.60 (d, / = 7.0 Hz, 2 H), 7.56-7.46 (m, 3 H), 4.36 (br s, 1 H), 4.04-3.72 (m, 6 H), 3.27-3.12 (m, 2 H), 2.51 (s, 3 H), 1.59 (dq, / = 13.8, 6.9 Hz, 2 H), 1.19 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.2 [M + H+] with a purity of >97%.
Propyl (5)-4-(4-chloro-2-phenylq lpiperazine-l-carboxylate (B018)
Figure imgf000197_0001
Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with (S)-n-propyl 2-methylpiperazine-l -carboxylate as reagent to afford B018.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.38 (s, 1 H), 8.29 (dd, / = 7.5, 3.3 Hz, 3 H), 8.11 (s, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.60-7.50 (m, 3 H), 4.25 (s, 1 H), 3.99 (pd, / = 10.6, 6.6 Hz, 3 H), 3.82 (d, / = 12.5 Hz, 1 H), 3.38-3.08 (m, 4 H), 1.66-1.51 (m, 2 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.1 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-phenyl ylpiperazine-l-carboxylate (B019)
Figure imgf000197_0002
Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine-l -carboxylate as reagent to afford B019.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.38 (s, 1 H), 8.32-8.23 (m, 3 H), 8.11 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.56 (q, / = 6.6 Hz, 3 H), 4.25 (s, 1 H), 3.99 (pd, / = 10.6, 6.5 Hz, 3 H), 3.82 (d, / = 12.6 Hz, 1 H), 3.36-3.06 (m, 4 H), 1.65-1.51 (m, 2 H), 1.14 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 452.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-3-methyl-2-(4-(trifluoromethyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B020)
Figure imgf000197_0003
Compound B020 was synthesized according to General Procedure I, K and CI using 4- trifluoromethylpropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.31 (d, / = 8.6 Hz, 1 H), 8.08 (d, / = 1.1 Hz, 1 H), 7.89 (dd, / = 19.3, 8.4 Hz, 4 H), 7.79 (dd, / = 8.6, 1.5 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.73-3.36 (m, 8 H), 2.51 (s, 3 H), 1.64-1.50 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 520.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(4-chlorophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate (B021)
Compound B021 was synthesized according to General Procedure I, K and CI using 4- chloropropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.29 (d, / = 8.6 Hz, 1 H), 8.07 (d, / = 1.3 Hz, 1 H), 7.77 (dd, / = 8.6, 1.5 Hz, 1 H), 7.64 (dd, / = 27.1 , 8.5 Hz, 4 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.79-3.36 (m, 8 H), 1.66- 1.49 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.1 [M + H+] with a purity of >96%.
4-(4-chloro-2-(4-fluorophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000198_0002
Compound B022 was synthesized according to General Procedure I, K and CI using 4- fluoropropiophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.27 (d, / = 8.6 Hz, 1 H), 8.04 (s, 1 H), 7.73 (d, / = 8.5 Hz, 1 H), 7.70-7.62 (m, 2 H), 7.33 (t, / = 8.9 Hz, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.62-3.37 (m, 8 H), 2.51 (s, 3 H), 1.70-1.52 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 470.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-3-methyl-2-(/7-tolyl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B023)
Figure imgf000198_0003
Compound B023 was synthesized according to General Procedure I, K and CI using 4- methylpropiophenone (General Procedure I) and (R)-n-propyl 3-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.26 (d, / = 8.6 Hz, 1 H), 7.99 (s, 1 H), 7.69 (d, / = 8.6 Hz, 1 H), 7.50 (d, / = 7.7 Hz, 2 H), 7.33 (d, / = 7.8 Hz, 2 H), 4.35 (s, 1 H), 4.08-3.68 (m, 5 H), 3.29-3.12 (m, 2 H), 3.02-2.89 (m, 1 H), 2.51 (s, 3 H), 2.41 (s, 3 H), 1.67-1.45 (m, 2H), 1.19 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 480.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-3-methyl-2-(/7-tolyl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B024)
Figure imgf000199_0001
Compound B024 was synthesized according to General Procedure I, K and CI using 4- methylpropiophenone (General Procedure I) and (S)-n-propyl 3-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.26 (d, / = 8.6 Hz, 1 H), 7.99 (s, 1 H), 7.69 (d, / = 8.7 Hz, 1 H), 7.50 (d, / = 7.9 Hz, 2 H), 7.33 (d, / = 7.8 Hz, 2 H), 4.35 (s, 1 H), 4.04-3.72 (m, 5 H), 3.30-2.90 (m, 3 H), 2.51 (s, 3 H), 2.41 (s, 3 H), 1.66-1.51 (m, 2 H), 1.19 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 480.2 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-3-methyl-2-(/7 erazine-l-carboxylate (B025)
Figure imgf000199_0002
Compound B025 was synthesized according to General Procedure I, K and CI using 4- methylpropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (d, / = 8.6 Hz, 1 H), 8.03 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.50 (d, / = 7.8 Hz, 2 H), 7.33 (d, / = 7.8 Hz, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.64-3.34 (m, 8 H), 2.51 (d, / = 5.1 Hz, 3 H), 2.41 (s, 3 H), 1.64-1.50 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 466.1 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(4-methoxyphenyl)-3-methylquinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B026)
Figure imgf000200_0001
Compound B026 was synthesized according to General Procedure I, K and CI using 4- methoxypropiophenone (General Procedure I) and (S)-n-propyl 3-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.24 (t, / = 19.6 Hz, 1 H), 8.01 (s, 1 H), 7.71 (d, / = 8.5 Hz, 1 H), 7.60 (d, / = 8.6 Hz, 2 H), 7.09 (d, / = 8.7 Hz, 2 H), 4.09-3.55 (m, 9 H), 3.26-2.79 (m, 3 H), 2.55 (s, 3 H), 1.65-1.48 (m, 2 H), 1.17 (s, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 496.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-3-methyl-2-(o- iperazine-l-carboxylate (B027)
Figure imgf000200_0002
Compound B027 was synthesized according to General Procedure I, K and CI using 2- methylpropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.30 (d, / = 8.6 Hz, 1 H), 8.05 (d, / = 1.2 Hz, 1 H), 7.78 (dd, / = 8.6, 1.5 Hz, 1 H), 7.46-7.30 (m, 3 H), 7.27 (d, / = 7.4 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.76-3.35 (m, 8 H), 2.30 (s, 3 H), 2.04 (s, 3 H), 1.65-1.49 (m, 2 H), 0.88 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 466.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-3-methyl-2-( -tolyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B028)
Figure imgf000200_0003
Compound B028 was synthesized according to General Procedure I, K and CI using 3- methylpropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.28 (d, / = 8.6 Hz, 1 H), 8.06 (d, / = 1.3 Hz, 1 H), 7.75 (dd, / = 8.6, 1.5 Hz, 1 H), 7.47-7.36 (m, 3 H), 7.33 (d, / = 6.5 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.75 - 3.34 (m, 8 H), 2.49 (s, 3 H), 2.41 (s, 3 H), 1.65-1.51 (m, 2H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 466.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-methoxyphenyl)-3-methylquinoline-7-carbonyl)piperazine-l- carboxylate (B029)
Compound B029 was synthesized according to General Procedure I, K and CI using 3- methoxypropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27 (d, / = 8.6 Hz, 1 H), 8.04 (s, 1 H), 7.73 (d, / = 8.6 Hz, 1 H), 7.43 (t, / = 7.8 Hz, 1 H), 7.14 (d, / = 7.0 Hz, 2 H), 7.07 (d, / = 9.9 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.83 (s, 3 H), 3.59-3.40 (m, 8 H), 2.50 (s, 3 H), 1.59 (dq, / = 14.2, 7.0 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >96%.
4-(4-chloro-2-(2,3-difluorophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-
Figure imgf000201_0002
Compound B030 was synthesized according to General Procedure I, K and CI using 2,3- difluoropropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.33 (d, / = 8.6 Hz, 1 H), 8.10 (s, 1 H), 7.82 (d, / = 8.7 Hz, 1 H), 7.70-7.55 (m, 1 H), 7.42 (dd, / = 9.6, 5.9 Hz, 2 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.77-3.50 (m, 8 H), 2.42 (s, 3 H), 1.64-1.52 (m, 2 H), 0.88 (dd, / = 13.4, 6.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 488.1 [M + H+] with a purity of >97%.
4-(4-chloro-2-(4-methoxyphenyl)-3-methylquinoline-7-carbonyl)piperazine-l-
Figure imgf000201_0003
Compound B031 was synthesized according to General Procedure I, K and CI using 4- methoxypropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.25 (d, / = 8.6 Hz, 1 H), 8.02 (s, 1 H), 7.70 (d, / = 8.6 Hz, 1 H), 7.57 (d, / = 8.5 Hz, 2H), 7.08 (d, / = 8.5 Hz, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.85 (s, 3 H), 3.75-3.42 (m, 8 H), 2.54 (s, 3 H), 1.64-1.51 (m, 2 H), 0.88 (dd, / = 13.2, 5.9 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >99%. 4-(4-chloro-2-(3-cyanophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000202_0001
Compound B032 was synthesized according to General Procedure I, K and CI using 3- cyanopropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.30 (d, / = 8.6 Hz, 1 H), 8.07 (s, 2 H), 7.95 (t, / = 6.5 Hz, 2 H), 7.79-7.68 (m, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.50 (d, / = 30.3 Hz, 8 H), 2.51 (s, 3 H), 1.65-1.51 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z All .1 [M + H+] with a purity of >99%.
Propyl 4-(2-(3-carbamoylphenyl -4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B033)
Figure imgf000202_0002
Compound B033 was synthesized according to General Procedure I, K and CI using 3- acetylbenzonitrile (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials. B033 was a side-product obtained from General Procedure K.
*H NMR (400 MHz, DMSO-<¾ δ 8.77 (s, 1 H), 8.57 (s, 1 H), 8.49 (d, / = 7.8 Hz, 1 H), 8.31 (d, / =
8.6 Hz, 1 H), 8.20-8.13 (m, 2 H), 8.04 (d, / = 7.8 Hz, 1 H), 7.77 (dd, / = 8.6, 1.6 Hz, 1 H), 7.67 (t, / =
7.7 Hz, 1 H), 7.53 (s, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.81-3.33 (m, 8 H), 1.65-1.50 (m, 2 H), 0.88 (dd, / = 14.6, 7.5 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-methoxyphenyl)-3-methylquinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B034)
Figure imgf000202_0003
Compound B034 was synthesized according to General Procedure I, K and CI using 4- methoxypropiophenone (General Procedure I) and (S)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.26 (d, / = 8.5 Hz, 1 H), 8.00 (s, 1 H), 7.70 (d, / = 8.5 Hz, 1 H), 7.58 (d, / = 8.4 Hz, 2 H), 7.08 (d, / = 8.4 Hz, 2 H), 4.41-3.69 (m, 8 H), 3.37-3.08 (m, 4 H), 2.54 (s, 3 H), 1.70-1.48 (m, 2 H), 1.12 (d, / = 6.4 Hz, 3 H), 0.97-0.76 (m, 3 H).
LCMS (ESI-TOF) nt/z 496.2 [M + H+] with a purity of >98%.
Propyl 4-(2-(3-carbamoylphenyl)-4-chloro-3-methylquinoline-7-carbonyl)piperazine-l- carboxylate (B035)
Figure imgf000203_0001
Compound B035 was synthesized according to General Procedure I, K and CI using 3- propionylbenzonitrile (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials. B035 was a side -product obtained from General Procedure K.
*H NMR (400 MHz, DMSO-<¾ δ 8.30 (d, / = 8.6 Hz, 1 H), 8.12 (s, 1 H), 8.09 (d, / = 1.3 Hz, 1 H), 8.07 (br s, 1 H), 8.02 (d, / = 7.9 Hz, 1 H), 7.82-7.72 (m, 2 H), 7.63 (t, / = 7.7 Hz, 1 H), 7.47 (br s, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.78-3.36 (m, 8 H), 2.52 (s, 3 H), 1.63-1.52 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 495.1 [M + H+] with a purity of >96%.
Propyl (/?)-4-(4-chloro-2-(4-methoxyphenyl)-3-methylquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate
Figure imgf000203_0002
Compound B036 was synthesized according to General Procedure I, K and CI using 4- methoxypropiophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.28 (d, / = 8.6 Hz, 1 H), 8.02 (s, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.60 (d, / = 8.5 Hz, 2 H), 7.10 (d, / = 8.6 Hz, 2 H), 4.33-3.72 (m, 8 H), 3.41-3.07 (m, 4 H), 2.56 (s, 3 H), 1.67-1.49 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.91 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.2 [M + H+] with a purity of >97%.
4-(4-chloro-2-(4-cyanophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000203_0003
B037 Compound B037 was synthesized according to General Procedure I, K and CI using 4- propionylbenzonitrile (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.31 (d, / = 8.6 Hz, 1 H), 8.09 (d, / = 1.0 Hz, 1 H), 8.03 (d, / = 8.3 Hz, 2 H), 7.85 (d, / = 8.4 Hz, 2 H), 7.79 (dd, / = 9.7, 2.6 Hz, 1 H), 3.97 (t, / = 6.5 Hz, 2 H), 3.76-3.46 (m, 8 H), 1.64-1.52 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z All .1 [M + H+] with a purity of >99%.
Propyl 4-(2-(3-(aminomethyl)phenyl)-4-chloro-3-methylquinoline-7-carbonyl)piperazine-l- carboxylate (B038)
Figure imgf000204_0001
Compound B038 was synthesized according to General Procedure I, K, CI and D using 3- propionylbenzonitrile (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.27 (d, / = 8.5 Hz, 1 H), 8.04 (s, 1 H), 7.73 (d, / = 8.7 Hz, 1 H), 7.55 (s, 1 H), 7.49-7.38 (m, 3 H), 3.99 (t, / = 6.4 Hz, 2 H), 3.83 (s, 2 H), 3.50 (d, / = 30.8 Hz, 8 H), 2.50 (d, / = 3.9 Hz, 3 H), 1.65-1.51 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.2 [M + H+] with a purity of >99%.
Propyl 4-(2-(3-(aminomethyl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B039)
Figure imgf000204_0002
Compound B039 was synthesized according to General Procedure I, K, CI and D using 3- acetylbenzonitrile (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.43-8.36 (m, 1 H), 8.26 (t, / = 8.0 Hz, 2 H), 8.13 (s, 2 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.55-7.45 (m, 2 H), 3.99 (t, / = 6.4 Hz, 2 H), 3.86 (s, 2 H), 3.62-3.40 (m, 8 H), 1.62-1.53 (m, 2 H), 0.90 (t, / = 7.5 Hz, 3 H).
LCMS (ESI-TOF) m/z 467.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(phenylamino)quinoline-7-carbonyl)piperazine-l-carboxylate (B040)
Figure imgf000205_0001
Step 1 : Phosphorus oxychloride (1.9 mL) was added to malonic acid (378.6 mg, 3.64 mmol, 1.1 equiv) at 0 °C. After 10 min, methyl 3-aminobenzoate (500 mg, 3.308 mmol) was added at the same temperature, before warming up to room temperature. The mixture was heated to reflux for 4 h before cooling it to room temperature. The contents were emptied into 2M aqueous sodium hydroxide and diluted with dichloromethane. At pH 7, the aqueous layer was extracted 4 times with dichloromethane, and the combined organic layers were washed with water and then brine. Upon drying over sodium sulfate, the mixture was filtered and concentrated. The crude material was purified by column chromatography to afford methyl 2,4-dichloroquinoline-7-carboxylate (112.2 mg, 13%).
Step 2: To a previously dried reaction vessel was added the above intermediate (28.5 mg, 0.11 mmol), aniline (25 mg, 0.139 mmol, 1.26 equiv), caesium carbonate (72 mg, 0.22 mmol, 2 equiv), XantPhos (19 mg, 0.0328 mmol, 0.3 equiv), tris(dibenzylideneacetone)dipalladium(0) (10 mg, 0.0109 mmol, 0.1 equiv) and previously degassed 1 ,4-dioxane (1 mL). The resulting mixture was heated at 110 °C for 2 h before filtering the contents with dichloromethane. The concentrated crude material was purified by column chromatography to afford methyl 4-chloro-2-(phenylamino)quinoline-7-carboxylate (30 mg, 87%).
Step 3: Methyl 4-chloro-2-(phenylamino)quinoline-7-carboxylate was hydrolysed according to General Procedure K to afford 4-chloro-2-(phenylamino)quinoline-7-carboxylic acid.
Step 4: Compound B040 was synthesized according to General Procedure CI using the above intermediate and n-propyl piperazine-l -carboxylate as reagents.
!H NMR (400 MHz, DMSO-<¾) δ 9.64 (s, 1 H), 8.03 (d, / = 8.4 Hz, 1 H), 7.94 (d, / = 7.7 Hz, 2 H), 7.73 (d, / = 1.3 Hz, 1 H), 7.40 (dd, / = 8.3, 1.5 Hz, 1 H), 7.38-7.33 (m, 2 H), 7.31 (s, 1 H), 7.01 (t, / = 7.3 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.75-3.33 (m, 8 H), 1.58 (dd, / = 13.8, 7.1 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 453.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-((4-methoxyphenyl)amino)quinoline-7-carbonyl)piperazine-l-carboxylate (B041)
Figure imgf000205_0002
Step 1 : To a previously dried reaction vessel was added the above intermediate methyl 2,4- dichloroquinoline-7-carboxylate (30 mg, 0.12 mmol), 4-methoxyaniline (18 mg, 0.1 mmol, 0.83 equiv), caesium carbonate (78 mg, 0.239 mmol, 2 equiv), XantPhos (20 mg, 0.0346 mmol, 0.29 equiv), tris(dibenzylideneacetone)dipalladium(0) (11 mg, 0.012 mmol, 0.1 equiv) and previously degassed 1 ,4-dioxane (1.5 mL). The resulting mixture was heated at 110 °C for 2 h before filtering the contents with dichloromethane. The concentrated crude material was purified by column chromatography to afford methyl 4-chloro-2-((4-methoxyphenyl)amino)quinoline-7-carboxylate (10 mg, 27%).
Step 2: Methyl 4-chloro-2-((4-methoxyphenyl)amino)quinoline-7-carboxylate was hydrolysed according to General Procedure K to afford 4-chloro-2-((4-methoxyphenyl)amino)quinoline-7- carboxylic acid.
Step 3: Compound B041 was synthesized according to General Procedure CI using the above intermediate and n-propyl piperazine-l-carboxylate as reagents.
*H NMR (400 MHz, DMSO-<¾) δ 9.48 (s, 1 H), 7.99 (d, / = 8.3 Hz, 1 H), 7.82 (d, / = 9.0 Hz, 2 H), 7.66 (d, / = 1.3 Hz, 1 H), 7.36 (dd, / = 8.3, 1.5 Hz, 1 H), 7.23 (s, 1 H), 6.94 (d, / = 9.1 Hz, 2 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.75 (s, 3 H), 3.71-3.35 (m, 8 H), 1.65-1.51 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 483.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(pyridin-3-ylamino)quinoline-7-carbonyl)piperazine-l-carboxylate (B042)
Figure imgf000206_0001
Step 1 : To a previously dried reaction vessel was added the above intermediate methyl 2,4- dichloroquinoline-7-carboxylate (30 mg, 0.12 mmol), 3-aminopyridine (14 mg, 0.144 mmol, 1.2 equiv), caesium carbonate (78 mg, 0.239 mmol, 2 equiv), XantPhos (20 mg, 0.0346 mmol, 0.29 equiv), tris(dibenzylideneacetone)dipalladium(0) (11 mg, 0.012 mmol, 0.1 equiv) and previously degassed 1,4-dioxane (1.5 mL). The resulting mixture was heated at 110 °C for 2 h before filtering the contents with dichloromethane. The concentrated crude material was purified by column chromatography to afford methyl 4-chloro-2-(pyridin-3-ylamino)quinoline-7-carboxylate (17 mg, 45%).
Step 2: Methyl 4-chloro-2-(pyridin-3-ylamino)quinoline-7-carboxylate was hydrolysed according to General Procedure K to afford 4-chloro-2-(pyridin-3-ylamino)quinoline-7-carboxylic acid.
Step 3: Compound B042 was synthesized according to General Procedure CI using the above intermediate and n-propyl piperazine-l-carboxylate as reagents.
*H NMR (400 MHz, DMSO-<¾) δ 9.87 (s, 1 H), 9.01 (d, / = 2.4 Hz, 1 H), 8.55-8.47 (m, 1 H), 8.22 (dd, / = 4.6, 1.3 Hz, 1 H), 8.06 (d, / = 8.4 Hz, 1 H), 7.79 (d, / = 1.2 Hz, 1 H), 7.44 (dd, / = 8.4, 1.5 Hz, 1 H), 7.38 (dd, / = 8.3, 4.7 Hz, 1 H), 7.34 (s, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.80-3.34 (m, 8 H), 1.64-1.51 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 454.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-3-methyl-2-(3-nitrophenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B043)
Figure imgf000206_0002
Compound B043 was synthesized according to General Procedure I, K and CI using 3- nitropropiophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.49-8.46 (m, 1 H), 8.38 (dd, / = 8.2, 2.3 Hz, 1 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.16-8.10 (m, 2 H), 7.85 (t, / = 8.0 Hz, 1 H), 7.80 (dd, / = 8.6, 1.5 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.81-3.32 (m, 8 H), 2.54 (s, 3 H), 1.67-1.51 (m, 2H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 497.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(3-aminophenyl)-4-chloro-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate (B044)
Figure imgf000207_0001
Compound B043 (39 mg, 0.08 mmol) was dissolved in ethanol (2 mL) and iron powder (27 mg, 0.48 mmol, 6 equiv) was added. Solid ammonium chloride (47 mg, 0.879 mmol, 11 equiv) was dissolved in water (0.5 mL) and the solution was added to the slurry. The slurry was heated at 70 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by preparative HPLC to afford compound B044 as an off-white solid upon lyophilization (6 mg, 16%).
*H NMR (400 MHz, DMSO-<¾) δ 8.25 (d, / = 8.6 Hz, 1 H), 8.01 (s, 1 H), 7.71 (d, / = 8.7 Hz, 1 H), 7.15 (t, / = 7.7 Hz, 1 H), 6.77 (s, 1 H), 6.69 (t, / = 7.0 Hz, 2 H), 5.04 (s, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.50 (d, / = 31.7 Hz, 8 H), 1.66-1.52 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 467.1 [M + H+] with a purity of >95%.
Propyl (/?)-4-(4-chloro-2-(4-methoxyphenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B045)
Figure imgf000207_0002
Compound B045 was synthesized according to General Procedure I, K and CI using 4- methoxyacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.33 (s, 1 H), 8.25 (t, / = 8.5 Hz, 3 H), 8.06 (s, 1 H), 7.68 (d, / = 8.7 Hz, 1 H), 7.11 (d, / = 8.9 Hz, 2 H), 4.37-3.74 (m, 10 H), 3.41-3.21 (m, 2 H), 1.60 (dt, / = 13.9, 6.9 Hz, 2 H), 1.14 (d, / = 5.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(4-metho erazine-l-carboxylate (B046)
Figure imgf000208_0001
Compound B045 was synthesized according to General Procedure I, K and CI using 4- methoxyacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.32 (s, 1 H), 8.25 (t, / = 9.3 Hz, 3 H), 8.08 (s, 1 H), 7.68 (d, / = 8.2 Hz, 1 H), 7.11 (d, / = 8.5 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.86 (s, 3 H), 3.51 (d, / = 30.8 Hz, 8 H), 1.58 (dt, / = 28.7, 14.4 Hz, 2 H), 0.90 (t, / = 7.5 Hz, 3 H).
LCMS (ESI-TOF) nt/z 468.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-methoxyphenyl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B047)
Figure imgf000208_0002
Compound B047 was synthesized according to General Procedure I, K and CI using 4- methoxyacetophenone (General Procedure I) and (5)-n -propyl 3-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.33 (s, 1 H), 8.30-8.21 (m, 3 H), 8.04 (s, 1 H), 7.66 (d, / = 8.5 Hz, 1 H), 7.11 (d, / = 8.9 Hz, 2 H), 4.57-3.68 (m, 10 H), 3.19 (d, / = 7.6 Hz, 2 H), 1.59 (dd, / = 14.2, 7.1 Hz, 2 H), 1.20 (d, / = 6.8 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 482.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3,5-difluo piperazine-l-carboxylate (B048)
Figure imgf000208_0003
Compound B048 was synthesized according to General Procedure I, K and CI using 3,5- difluoroacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure C2) as reagents.
*H NMR (400 MHz, DMSO-<¾ δ 8.61 (s, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.16 (d, / = 23.4 Hz, 1 H), 8.09 (d, / = 7.0 Hz, 1 H), 7.80 (d, / = 8.5 Hz, 1 H), 7.45 (t, / = 9.0 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.83-3.33 (m, 8 H), 1.59 (dd, / = 13.8, 6.9 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 474.1 [M + H+] with a purity of >98%. Propyl 4-(4-chloro-2-(3-fluorophen l)quinoline-7-carbonyl)piperazine-l-carboxylate (B049)
Figure imgf000209_0001
Compound B049 was synthesized according to General Procedure I, K and CI using 3- fluoroacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
!H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.44 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.20-8.04 (m, 3 H), 7.75 (dd, / = 8.5, 1.4 Hz, 1 H), 7.60 (dd, / = 14.1 , 8.0 Hz, 1 H), 7.35 (td, / = 8.2, 2.0 Hz, 1 H), 3.99 (t, J = 6.6 Hz, 2 H), 3.52 (d, / = 30.8 Hz, 8 H), 1.68-1.52 (m, 2H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 456.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(pyrrolidin-l-yl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B050)
Figure imgf000209_0002
Compound B050 was synthesized according to General Procedure I, K and CI using 4'-(.l - pyrrolidino)aeetOphenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.22 (s, 1 H), 8.16 (dd, / = 8.5, 6.9 Hz, 3 H), 7.99 (s, 1 H), 7.59 (d, / = 9.9 Hz, 1 H), 6.68 (d, / = 8.8 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.51 (d, / = 31.8 Hz, 8 H), 3.34 (d, / = 6.4 Hz, 4 H), 2.00 (t, / = 6.4 Hz, 4 H), 1.60 (dd, / = 14.1 , 6.9 Hz, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 507.2 [M + H+] with a purity of >96%.
Propyl 4-(4-chloroquinoline-7-carbo xylate (B051)
Figure imgf000209_0003
Compound B051 was synthesized via General Procedure C2 using n-propyl piperazine-l-carboxylate and 4-chloroquinoline-6-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.92 (d, / = 4.7 Hz, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.12 (s, 1 H), 7.86 (d, / = 4.7 Hz, 1 H), 7.79 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.37 (m, 8 H), 1.72-1.45 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 362.1 [M + H+] with a purity of >96%. Propyl ( ?)-4-(4-chloro-2-(p-tolyl)quinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate (B052)
Figure imgf000210_0001
Compound B052 was synthesized according to General Procedure I, K and C2 using 4- methylacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine (General Procedure C2) as reagents.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.19 (d, / = 8.2 Hz, 2 H), 8.09 (d, / = 1.1 Hz, 1 H), 7.70 (dd, / = 8.5, 1.5 Hz, 1 H), 7.37 (d, / = 8.0 Hz, 2 H), 4.37-3.71 (m, 6 H), 3.44-3.10 (m, 3 H), 2.40 (s, 3 H), 1.72-1.47 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 466.1 [M + H+] with a purity of >97%.
Propyl ( ?)-4-(2-(3-carbamoylphenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B053)
Figure imgf000210_0002
Compound B053 was synthesized according to General Procedure I, K and CI using 3- acetylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1 -carboxylate (General Procedure CI) as starting materials. B053 was a side -product obtained from General Procedure K.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.73 (s, 1 H), 8.47 (s, 1 H), 8.45 (d, / = 7.9 Hz, 1 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.15 (s, 1 H), 8.03 (d, / = 7.7 Hz, 1 H), 7.77-7.73 (m, 1 H), 7.64 (t, / = 7.7 Hz, 1 H), 4.40-3.71 (m, 6 H), 3.38-3.10 (m, 3 H), 1.67-1.50 (m, 2H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.1 [M + H+] with a purity of >96%.
Propyl (/?)-4-(2-(3-(aminomethyl)phenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B054)
Figure imgf000210_0003
Compound B054 was synthesized according to General Procedure I, K, CI and D using 3- acetylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1 -carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.38 (s, 1 H), 8.30-8.22 (m, 2 H), 8.15-8.07 (m, 2 H), 7.72 (dd, / = 8.5, 1.5 Hz, 1 H), 7.53-7.44 (m, 2 H), 4.34-3.74 (m, 7 H), 3.43-3.05 (m, 4 H), 1.66-1.51 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 481.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(dimethylamino)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B055)
Figure imgf000211_0001
Compound B055 was synthesized according to General Procedure I, K and CI using 4' - dimethylaminoacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.34 (s, 1 H), 8.20 (dd, / = 8.6, 4.7 Hz, 3 H), 8.01 (s, 1 H), 7.63 (d, / = 8.5 Hz, 1 H), 6.84 (d, / = 9.0 Hz, 2 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.79-3.35 (m, 8 H), 3.03 (s, 6 H), 1.67-1.50 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 481.2 [M + H+] with a purity of >95%.
Propyl (/?)-4-(2-(3-aminophenyl)-4-chloro-3-methylquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B056)
Figure imgf000211_0002
Step 1 : Intermediate 2 for compound B043 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine-l -carboxylate to afford propyl (R)-4-(4-chloro-3-methyl-2-(3- nitrophenyl)quinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate.
Step 2: Intermediate from Step 1 (387 mg, 0.78 mmol) was dissolved in ethanol (20 mL) and iron powder (263 mg, 4.7 mmol, 6 equiv) was added. Solid ammonium chloride (460.1 mg, 8.6 mmol, 11 equiv) was dissolved in water (5 mL) and the solution was added to the slurry. The slurry was heated at 70 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by preparative HPLC to afford compound B056 as an off-white solid upon lyophilization (221 mg, 59%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (d, / = 8.6 Hz, 1 H), 7.99 (d, / = 1.1 Hz, 1 H), 7.70 (dd, / = 8.6, 1.5 Hz, 1 H), 7.15 (t, / = 7.8 Hz, 1 H), 6.77 (d, / = 1.7 Hz, 1 H), 6.70 (dd, / = 9.8, 4.8 Hz, 2 H), 5.03 (s, 2 H), 4.33-3.71 (m, 6 H), 3.43-3.09 (m, 3 H), 2.50 (s, 3 H), 1.65-1.46 (m, 2 H), 1.09 (t, / = 16.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 481.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(4-ethynylphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B057)
Figure imgf000212_0001
Compound B057 was synthesized according to General Procedure I, K and CI using 4- acetylphenylacetylene (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.53 (s, 1 H), 8.36 (d, / = 8.4 Hz, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.15 (s, 1 H), 7.77 (dd, / = 8.5, 1.4 Hz, 1 H), 7.68 (d, / = 8.4 Hz, 2 H), 4.39 (s, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.82-3.35 (m, 8 H), 1.63-1.52 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 462.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-(dimethylamino)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B058)
Figure imgf000212_0002
Compound B058 was synthesized according to General Procedure I, K and CI using 3'- dimethylaminoacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.45 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.14 (s, 1 H), 7.73 (dd, / = 8.5, 1.4 Hz, 1 H), 7.62 (s, 1 H), 7.59 (d, / = 7.7 Hz, 1 H), 7.37 (t, / = 7.9 Hz, 1 H), 6.91 (dd, / = 8.2, 2.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.82-3.34 (m, 8 H), 3.02 (s, 6 H), 1.63-1.51 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-ethynylphenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B059)
Figure imgf000212_0003
Compound B059 was synthesized according to General Procedure I, K and CI using 4- acetylphenylacetylene (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.42 (s, 1 H), 8.32 (d, / = 8.4 Hz, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.12 (d, / = 0.9 Hz, 1 H), 7.74 (dd, / = 8.5, 1.4 Hz, 1 H), 7.65 (d, / = 8.4 Hz, 2 H), 4.36-3.68 (m, 7 H), 3.42-3.09 (m, 3 H), 1.67-1.52 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 476.1 [M + H+] with a purity of >96%. Propyl (/?)-4-(4-chloro-2-(4-(dimethylamino)phenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B060)
Figure imgf000213_0001
Compound B060 was synthesized according to General Procedure I, K and CI using 4' - dimethylaminoacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l - carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.23 (s, 1 H), 8.22-8.10 (m, 3 H), 7.99 (s, 1 H), 7.60 (d, / = 8.5 Hz, 1 H), 6.84 (d, / = 9.0 Hz, 2 H), 4.46-3.65 (m, 6 H), 3.37-3.09 (m, 3 H), 3.02 (s, 6 H), 1.59 (dt, / = 13.9, 7.1 Hz, 2 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(3-(dimethylamino)phenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B061)
Figure imgf000213_0002
Compound B061 was synthesized according to General Procedure I, K and C I using 3 '- dimethylaminoacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l - carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.34 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.10 (s, 1 H), 7.70 (dd, / = 8.5, 1.5 Hz, 1 H), 7.60 (s, 1 H), 7.54 (d, / = 7.7 Hz, 1 H), 7.35 (t, / = 7.9 Hz, 1 H), 6.90 (dd, / = 8.3, 2.2 Hz, 1 H), 4.38-3.70 (m, 6 H), 3.42-3.11 (m, 3 H), 3.01 (s, 6 H), 1.66-1.54 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(4-cyanophenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B062)
Figure imgf000213_0003
Compound B062 was synthesized according to General Procedure I, K and CI using 4- acetylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l -carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.49 (d, / = 8.8 Hz, 3 H), 8.31 (d, / = 8.6 Hz, 1 H), 8.15 (d, / = 1.0 Hz, 1 H), 8.00 (d, / = 8.4 Hz, 2 H), 7.78 (dd, / = 8.5, 1.5 Hz, 1 H), 4.50-3.56 (m, 6 H), 3.43- 3.09 (m, 3 H), 1.67-1.52 (m, 2 H), 1.14 (d, / = 6.1 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 477.1 [M + H+] with a purity of >95%.
Propyl (/?)-4-(2-(4-carbamoylphenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B063)
Figure imgf000214_0001
Compound B063 was synthesized according to General Procedure I, K and CI using 4- acetylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l -carboxylate (General Procedure CI) as starting materials. Compound B063 is a side -product in the synthesis of B062.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.45 (s, 1 H), 8.36 (d, / = 7.9 Hz, 2 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.14 (s, 1 H), 8.05 (d, / = 7.9 Hz, 2 H), 7.75 (d, / = 8.5 Hz, 1 H), 4.46-3.67 (m, 6 H), 3.47-3.10 (m, 3 H), 1.59 (dt, / = 14.1 , 7.1 Hz, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 495.1 [M + H+] with a purity of >95%.
Propyl (/?)-4-(2-(4-(aminomethyl)phenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B064)
Figure imgf000214_0002
Compound B064 was synthesized from B062 using General Procedure D.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.23 (d, / = 8.3 Hz, 2 H), 8.09 (s, 1 H), 7.74-7.62 (m, 1 H), 7.52 (d, / = 8.2 Hz, 2 H), 4.36-3.74 (m, 8 H), 3.39-3.14 (m, 3 H), 1.67-1.53 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 481.2 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(4-fluoroph erazine-l-carboxylate (B065)
Figure imgf000214_0003
Compound B065 was synthesized according to General Procedure I, K and CI using 4- fluoroacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.39 (s, 1 H), 8.36 (dd, / = 8.8, 5.6 Hz, 2 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.12 (s, 1 H), 7.73 (dd, / = 8.5, 1.4 Hz, 1 H), 7.36 (t, / = 8.8 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.51 (d, / = 30.7 Hz, 8 H), 1.67-1.51 (m, 2 H), 0.96-0.80 (m, 3 H).
LCMS (ESI-TOF) nt/z 456.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(trifluoromethyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B066)
Figure imgf000215_0001
Compound B066 was synthesized according to General Procedure I, K and CI using 4- trifluoroacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.51 (d, / = 8.1 Hz, 2 H), 8.48 (s, 1 H), 8.31 (d, / = 8.5 Hz, 1 H), 8.17 (s, 1 H), 7.90 (d, / = 8.2 Hz, 2 H), 7.78 (d, / = 8.5 Hz, 1 H), 4.00 (t, / = 6.5 Hz, 2 H), 3.52 (d, / = 31.9 Hz, 8 H), 1.68-1.51 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 506.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-ethylphe ne-l-carboxylate (B067)
Figure imgf000215_0002
Compound B067 was synthesized according to General Procedure I, K and CI using 3- ethylacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.48 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.15 (t, / = 10.8 Hz, 3 H), 7.74 (dd, / = 8.5, 1.4 Hz, 1 H), 7.49 (t, / = 7.6 Hz, 1 H), 7.40 (d, / = 7.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.83-3.34 (m, 8 H), 2.75 (q, / = 7.6 Hz, 2 H), 1.59 (dd, / = 13.9, 6.9 Hz, 2 H), 1.28 (t, / = 7.6 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 466.2 [M + H+] with a purity of >99%.
4-(4-chloro-2-(4-fluoro-3-methylphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000215_0003
Compound B068 was synthesized according to General Procedure I, K and CI using 4-fluoro-3- methylacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.48 (s, 1 H), 8.29 (t, / = 8.4 Hz, 2 H), 8.24-8.17 (m, 1 H), 8.13 (d, / = 0.9 Hz, 1 H), 7.74 (dd, / = 8.5, 1.5 Hz, 1 H), 7.33 (t, / = 9.1 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.74-3.34 (m, 8 H), 2.37 (s, 3 H), 1.59 (dd, / = 13.8, 6.7 Hz, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 470.1 [M + H+] with a purity of >96%.
Propyl (/?)-4-(4-chloro-2-(3-morpholinophenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B069)
Figure imgf000216_0001
Compound B069 was synthesized according to General Procedure I, K and CI using l-(3-morpholin- 4-yl-phenyl)ethanone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.39 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 7.82 (s, 1 H), 7.76-7.67 (m, 2 H), 7.41 (t, / = 7.9 Hz, 1 H), 7.10 (d, / = 8.0 Hz, 1 H), 4.42-3.70 (m, 10 H), 3.40-3.13 (m, 7 H), 1.65-1.54 (m, 2 H), 1.13 (s, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 537.2 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-ethynylph nyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B070)
Figure imgf000216_0002
Compound B070 was synthesized according to General Procedure I, K and CI using 3- acetylphenylacetylene (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.56 (s, 1 H), 8.45 (s, 1 H), 8.37 (d, / = 7.5 Hz, 1 H), 8.29 (d, / = 8.3 Hz, 1 H), 8.18 (s, 1 H), 7.77 (d, / = 7.9 Hz, 1 H), 7.70-7.53 (m, 2 H), 4.31 (s, 1 H), 3.98 (t, / = 6.2 Hz, 2 H), 3.81-3.36 (m, 8 H), 1.59 (dd, / = 11.0, 4.8 Hz, 2 H), 0.89 (t, / = 6.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 462.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3-ethynylphenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B071)
Figure imgf000216_0003
B071 Compound B071 was synthesized according to General Procedure I, K and CI using 3- acetylphenylacetylene (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.46 (s, 1 H), 8.41 (s, 1 H), 8.33 (d, / = 7.7 Hz, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.14 (s, 1 H), 7.74 (dd, / = 8.5, 1.4 Hz, 1 H), 7.60 (dt, / = 15.3, 7.6 Hz, 2 H), 4.35- 3.70 (m, 7 H), 3.41-3.10 (m, 3 H), 1.68-1.52 (m, 2 H), 1.14 (d, / = 5.3 Hz, 3 H), 0.93-0.85 (m, 3 H).
LCMS (ESI-TOF) nt/z 476.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-nitrophe zine-l-carboxylate (B072)
Figure imgf000217_0001
Compound B072 was synthesized according to General Procedure I, K and CI using 4- nitroacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.56 (d, / = 8.9 Hz, 2 H), 8.53 (s, 1 H), 8.38 (d, / = 8.9 Hz, 2 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.19 (d, / = 0.9 Hz, 1 H), 7.80 (dd, / = 8.5, 1.5 Hz, 1 H), 4.00 (t, / = 6.6 Hz, 2 H), 3.67-3.38 (m, 8 H), 1.68-1.47 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 483.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(4-aminophenyl)-4-c razine-l-carboxylate (B073)
Figure imgf000217_0002
Compound B072 (45 mg, 0.093 mmol) was dissolved in ethanol (5 mL) and iron powder (45 mg, 0.806 mmol, 8.6 equiv) was added. Solid ammonium chloride (45 mg, 0.841 mmol, 9 equiv) was dissolved in water (5 mL) and the solution was added to the slurry. The slurry was heated at 80 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by column chromatography to afford compound B073 as a yellow solid upon lyophilization (25 mg, 59%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.21-8.14 (m, 2 H), 8.02 (d, / = 8.6 Hz, 2 H), 7.98 (s, 1 H), 7.60 (d, / = 8.5 Hz, 1 H), 6.71 (d, / = 8.6 Hz, 2 H), 5.45 (s, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.62-3.40 (m, 8 H), 1.66-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 453.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(2-(4-aminophenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B074)
Step 1 : Intermediate 2 for compound B072 was subjected to General Procedure CI with (R)-n-propyl 2-methylpiperazine-l -carboxylate to afford propyl (R)-4-(4-chloro-2-(4-nitrophenyl)quinoline-7- carbonyl)-2-methylpiperazine- 1 -carboxylate.
Step 2: Intermediate from Step 1 (70 mg, 0.141 mmol) was dissolved in ethanol (5 mL) and iron powder (70 mg, 1.25 mmol, 8.9 equiv) was added. Solid ammonium chloride (70 mg, 1.31 mmol, 9.3 equiv) was dissolved in water (5 mL) and the solution was added to the slurry. The slurry was heated at 80 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by column chromatography to afford compound B074 as a yellow solid upon lyophilization (36.2 mg, 55%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27-8.08 (m, 2 H), 8.02 (d, / = 8.5 Hz, 2 H), 7.96 (s, 1 H), 7.59 (d, / = 8.5 Hz, 1 H), 6.71 (d, / = 8.6 Hz, 2 H), 5.46 (s, 2 H), 4.46-3.62 (m, 6 H), 3.36-3.10 (m, 3 H), 1.67-1.52 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 467.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-(pyrrolidin-l-yl)phenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B075)
Figure imgf000218_0002
Compound B075 was synthesized according to General Procedure I, K and CI using 4'-(l- pyrrolidino)acetophenone (General Procedure I) and (R)-n -propyl 2-methylpiperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.25-8.10 (m, 4 H), 7.97 (d, / = 1.0 Hz, 1 H), 7.59 (dd, / = 8.5, 1.4 Hz, 1 H), 6.68 (d, / = 8.9 Hz, 2 H), 4.35-3.68 (m, 6 H), 3.35 (t, / = 6.5 Hz, 4 H), 3.32-3.08 (m, 3 H), 2.00 (t, / = 6.5 Hz, 4 H), 1.66-1.54 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-(dimethylamino)phenyl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B076)
Figure imgf000218_0003
B076 Compound B076 was synthesized according to General Procedure I, K and CI using 4'- dimethylaminoacetophenone (General Procedure I) and (S)-n-propyl 3-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27-8.11 (m, 4 H), 7.97 (d, / = 1.1 Hz, 1 H), 7.58 (dd, / = 8.5, 1.6 Hz, 1 H), 6.84 (d, / = 8.6 Hz, 2 H), 4.49-3.69 (m, 6 H), 3.32-3.14 (m, 2 H), 3.02 (s, 6 H), 3.02-2.92 (m, 1 H), 1.67-1.53 (m, 2 H), 1.20 (d, / = 6.8 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.2 [M + H+] with a purity of >95%.
Propyl 4-(2-(3-aminophenyl)-4-c razine-l-carboxylate (B077)
Figure imgf000219_0001
B077
Step 1-3: Propyl 4-(4-chloro-2-(3-nitrophenyl)quinoline-7-carbonyl)piperazine-l-carboxylate was synthesized via General Procedure I, K and CI using 3-nitroacetophenone (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
Step 4: Intermediate from above (200 mg, 0.414 mmol) was dissolved in ethanol (6 mL) and iron powder (138.77 mg, 2.485 mmol, 6 equiv) was added. Solid ammonium chloride (243.7 mg, 4.56 mmol, 11 equiv) was dissolved in water (5 mL) and the solution was added to the slurry. The slurry was heated at 70 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by column chromatography to afford compound B077 as an off-white solid upon lyophilization (180 mg, 96%).
*H NMR (400 MHz, DMSO-<¾) δ 8.27 (s, 1 H), 8.26 (d, / = 6.3 Hz, 1 H), 8.09 (s, 1 H), 7.73 (d, / = 8.5 Hz, 1 H), 7.52 (s, 1 H), 7.40 (d, / = 7.6 Hz, 1 H), 7.20 (t, / = 7.8 Hz, 1 H), 6.74 (d, / = 7.5 Hz, 1 H), 5.29 (s, 2 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.82-3.33 (m, 8 H), 1.59 (dd, / = 13.9, 6.9 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 453.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(2-(3-aminophenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B078)
Figure imgf000219_0002
Step 1-3: Propyl (R)-4-(4-chloro-2-(3-nitrophenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylatewas synthesized via General Procedure I, K and CI using 3-nitroacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
Step 4: Intermediate from above (300 mg, 0.604 mmol) was dissolved in ethanol (8 mL) and iron powder (202.3 mg, 3.622 mmol, 6 equiv) was added. Solid ammonium chloride (355.3 mg, 6.64 mmol, 11 equiv) was dissolved in water (5 mL) and the solution was added to the slurry. The slurry was heated at 70 °C for 15 min and then diluted with methanol and filtered upon cooling. The concentrated crude material was purified by column chromatography to afford compound B078 as an off-white solid upon lyophilization (180 mg, 64%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (d, / = 8.6 Hz, 1 H), 8.20 (s, 1 H), 8.06 (s, 1 H), 7.70 (d, / = 8.6 Hz, 1 H), 7.52 (s, 1 H), 7.39 (d, / = 7.6 Hz, 1 H), 7.20 (t, / = 7.8 Hz, 1 H), 6.75 (d, / = 7.4 Hz, 1 H), 5.08 (s, 2 H), 4.35-3.69 (m, 6 H), 3.41-3.10 (m, 3 H), 1.69-1.52 (m, 2 H), 1.13 (d, / = 5.9 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 467.1 [M + H+] with a purity of >98%.
( ?)-4-(4-chloro-2-cyclopropylquinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate
Figure imgf000220_0001
B079
Compound B079 was synthesized via General Procedure L using propyl (R)-4-(2,4-dichloroquinoline- 7-carbonyl)-2-methylpiperazine-l-carboxylate and cyclopropyl boronic acid as starting material.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.17 (d, / = 8.5 Hz, 1 H), 7.87 (d, / = 1.1 Hz, 1 H), 7.71 (s, 1 H), 7.60 (dd, / = 8.5, 1.5 Hz, 1 H), 4.35-3.68 (m, 6 H), 3.31-3.09 (m, 3 H), 2.43-2.26 (m, 1 H), 1.70- 1.51 (m, 2 H), 1.20-1.03 (m, 7 H), 0.93-0.85 (m, 3 H).
LCMS (ESI-TOF) m/z 416.1 [M + H+] with a purity of >98%.
( ?)-4-(4-chloro-2-(thiazol-2-yl)quinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate
Figure imgf000220_0002
Compound B080 was synthesized according to General Procedure I, K and CI using 2-acetylthiazole (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.44 (s, 1 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.11 (d, / = 0.9 Hz, 1 H), 8.09 (d, / = 3.1 Hz, 1 H), 7.97 (d, / = 3.1 Hz, 1 H), 7.78 (dd, / = 8.6, 1.5 Hz, 1 H), 4.39-3.67 (m, 6 H), 3.52-3.11 (m, 3 H), 1.69-1.50 (m, 2 H), 1.14 (d, / = 6.1 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.0 [M + H+] with a purity of >99%.
Propyl 3-(4-chloro-2-phenylquinoline-7-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-9-carboxylate (B081)
Figure imgf000221_0001
Step 1 : Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with tert-butyl 3,9-diazabicyclo[3.3.1]nonane-9-carboxylate as reagent to afford tert- butyl 3-(4-chloro-2-phenylquinoline-7-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-9-carboxylate.
Step 2: Intermediate from Step 1 (118.5 mg, 0.2408 mmol) was dissolved in dichloromethane (0.2 mL) and trifluoroacetic acid was added (0.2 mL). After 20 min, the mixture was concentrated and re- dissolved in ethyl acetate. The organic layer was washed with saturated bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated to give crude (3,9-diazabicyclo[3.3.1]nonan-3- yl)(4-chloro-2-phenylquinolin-7-yl)methanone.
Step 3: The crude intermediate from above (77.7 mg, 0.1982 mmol) was dissolved in dichloromethane (2 mL) and triethylamine (60 μL·, 0.43 mmol, 2.2 equiv) and propyl chloroformate (30 μL·, 0.258 mmol, 1.3 equiv) were added. After 30 min, the mixture was quenched by adding saturated ammonium chloride, followed by extraction with dichloromethane. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography to afford B081 (27.7 mg, 29%) as a white solid upon lyophilization.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.38 (s, 1 H), 8.32-8.24 (m, 3 H), 8.07 (d, / = 1.1 Hz, 1 H), 7.69 (dd, / = 8.5, 1.6 Hz, 1 H), 7.63-7.48 (m, 3 H), 4.52-3.82 (m, 5 H), 3.54-3.15 (m, 2 H), 2.20- 2.04 (m, 1 H), 2.00-1.50 (m, 8 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 478.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(4-(dimethylcarbamoyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylat
Figure imgf000221_0002
Compound B082 was synthesized according to General Procedure I, K and CI using 4-acetyl-N,N- dimethylbenzamide (General Procedure I) and (R)-n -propyl 2-methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.42 (d, / = 8.1 Hz, 1 H), 8.35 (d, / = 8.3 Hz, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.13 (s, 1 H), 7.74 (dd, / = 8.5, 1.4 Hz, 1 H), 7.58 (d, / = 8.3 Hz, 2 H), 4.40-3.73 (m, 6 H), 3.40-3.10 (m, 3 H), 2.99 (s, 6 H), 1.68-1.54 (m, 2 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 523.2 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-(3-(methylamino)phenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B083)
Figure imgf000222_0001
Compound B083 was synthesized according to General Procedure I, K and CI using l -(3- (methylamino)phenyl)ethanone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (d, / = 8.7 Hz, 1 H), 8.25 (s, 1 H), 8.08 (s, 1 H), 7.70 (dd, / = 8.5, 1.5 Hz, 1 H), 7.49-7.36 (m, 2 H), 7.26 (t, / = 7.8 Hz, 1 H), 6.72 (d, / = 8.0 Hz, 1 H), 5.61 (d, / = 5.1 Hz, 1 H), 4.34-3.75 (m, 6 H), 3.39-3.09 (m, 3 H), 2.80 (d, / = 5.1 Hz, 3 H), 1.67-1.49 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(thiophen-2- azine-l-carboxylate (B084)
Figure imgf000222_0002
Compound B084 was synthesized according to General Procedure I, K and CI using 2- acetylthiophene (General Procedure I) and n-propyl piperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.49 (s, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 8.16 (d, / = 3.5 Hz, 1 H), 8.02 (s, 1 H), 7.81 (d, / = 5.1 Hz, 1 H), 7.70 (d, / = 8.4 Hz, 1 H), 7.30-7.18 (m, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.78-3.34 (m, 8 H), 1.59 (d, / = 6.8 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 444.0 [M + H+] with a purity of >95%.
Propyl (/?)-4-(4-chloro-2-(thiophen-2-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B085)
Figure imgf000222_0003
Intermediate from General Procedure K of the synthesis of B084 was reacted with (R)-n-propyl 2- methylpiperazine-l-carboxylic acid using General Procedure CI to give compound B085.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.23 (d, / = 8.5 Hz, 1 H), 8.07 (d, / = 2.8 Hz, 1 H), 7.98 (s, 1 H), 7.76 (d, / = 5.0 Hz, 1 H), 7.67 (dd, / = 8.5, 1.5 Hz, 1 H), 7.23 (dd, / = 5.0, 3.8 Hz, 1 H), 4.36-3.67 (m, 6 H), 3.39-3.08 (m, 3 H), 1.67-1.51 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 458.1 [M + H+] with a purity of >95%. Propyl (/?)-4-(4-chloro-2-(l-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B086)
Figure imgf000223_0001
Compound B086 was synthesized according to General Procedure I, K and CI using 1 -(1 -methyl- 1H- indazol-5-yl)ethanone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l -carboxylate (General Procedure CI) as starting materials.
!H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.47 (s, 1 H), 8.41 (dd, / = 8.9, 1.6 Hz, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.17 (s, 1 H), 8.11 (d, / = 1.0 Hz, 1 H), 7.77 (d, / = 8.9 Hz, 1 H), 7.70 (dd, / = 8.5, 1.5 Hz, 1 H), 4.45-3.71 (m, 9 H), 3.43-3.13 (m, 3 H), 1.60 (dd, / = 14.0, 6.7 Hz, 2 H), 1.15 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 506.2 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(4-fluoro-3-methoxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B087)
Figure imgf000223_0002
B087
Compound B087 was synthesized according to General Procedure I, K and CI using 4-fluoro-3- methoxyacetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.45 (s, 1 H), 8.27 (d, / = 8.4 Hz, 1 H), 8.13 (d, / = 1.0 Hz, 1 H), 8.06 (dd, / = 8.5, 2.0 Hz, 1 H), 7.94-7.87 (m, 1 H), 7.73 (dd, / = 8.5, 1.5 Hz, 1 H), 7.36 (dd, / = 11.1 , 8.6 Hz, 1 H), 4.06-3.95 (m, 5 H), 3.66-3.39 (m, 8 H), 1.66-1.49 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-fluoro-3-methoxyphenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B088)
Figure imgf000223_0003
Compound B088 was synthesized according to CI between intermediate K from synthesis of B087 and (R)-n-propyl 2-methylpiperazine-l-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.44 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.11 (d, / = 1.0 Hz, 1 H), 8.06 (dd, / = 8.4, 2.1 Hz, 1 H), 7.91 (ddd, / = 8.4, 4.4, 2.1 Hz, 1 H), 7.72 (dd, / = 8.5, 1.5 Hz, 1 H), 7.35 (dd, / = 11.2, 8.5 Hz, 1 H), 4.36-3.75 (m, 9 H), 3.39-3.11 (m, 3 H), 1.66-1.54 (m, 2 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 500.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(2-(4-(lH-imidazol-l-yl)phenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate
Figure imgf000224_0001
Compound B089 was synthesized according to General Procedure I, K and CI using 4-(imidazol-l - yl)acetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.46 (s, 1 H), 8.45 (d, / = 8.8 Hz, 2 H), 8.31 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.13 (s, 1 H), 7.83 (d, / = 8.6 Hz, 2 H), 7.79 (s, 1 H), 7.73 (dd, / = 8.5, 1.4 Hz, 1 H), 7.14 (s, 1 H), 4.38-3.61 (m, 6 H), 3.40-3.10 (m, 3 H), 1.66-1.51 (m, 2 H), 1.15 (d, / = 6.4 Hz, 3 H), 0.90 (t, / = 6.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 518.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-morpholinophenyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B090)
Figure imgf000224_0002
Compound B090 was synthesized according to General Procedure I, K and CI using 4- morpholinoacetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27 (s, 1 H), 8.20 (t, / = 8.3 Hz, 3 H), 8.02 (s, 1 H), 7.63 (dd, / = 8.5, 1.3 Hz, 1 H), 7.07 (d, / = 8.9 Hz, 2 H), 4.37-3.72 (m, 10 H), 3.36-3.23 (m, 5 H), 3.16 (dd, / = 27.3, 13.6 Hz, 2 H), 1.66-1.52 (m, 2 H), 1.14 (d, / = 6.5 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 537.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(l-methyl-lH-pyrazol-5-yl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B091)
Figure imgf000224_0003
B091 Compound B091 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3 -methylpiperazine- 1 -carboxylate and 1 -methylpyrazole-5 -boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27 (d, / = 8.5 Hz, 1 H), 8.21 (s, 1 H), 8.11 (d, / = 1.1 Hz, 1 H), 7.73 (dd, / = 8.5, 1.6 Hz, 1 H), 7.54 (d, / = 2.0 Hz, 1 H), 7.11 (d, / = 2.0 Hz, 1 H), 4.40-4.36 (m, 1 H), 4.33 (s, 3 H), 4.09-3.74 (m, 5 H), 3.30-2.93 (m, 3 H), 1.67-1.53 (m, 2 H), 1.21 (d, / = 6.8 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H). LCMS (ESI-TOF) m/z 456.1 [M + H+] with a purity of >99%.
Propyl 4-(2-(4-carbamoylphenyl)-4-chloro-3-methylquinoline-7-carbonyl)piperazine-l- carboxylate (B092)
Figure imgf000225_0001
Compound B092 was synthesized according to General Procedure I, K and CI using 4- propanoylbenzonitrile (General Procedure I) and n-propyl piperazine-1 -carboxylate (General Procedure CI) as starting materials. Compound B092 is a side product of the synthesis of propyl 4-(4- chloro-2-(4-cyanophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.29 (d, / = 8.6 Hz, 1 H), 8.06 (s, 1 H), 8.01 (d, / = 8.3 Hz, 2 H), 7.76-7.72 (m, 1 H), 7.68 (d, / = 8.3 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.62-3.35 (m, 8 H), 2.51 (s, 3 H), 1.78-1.47 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(4-(aminomethyl)phenyl)-4-chloro-3-methylquinoline-7-carbonyl)piperazine-l- carboxylate (B093)
Figure imgf000225_0002
Compound B093 was synthesized according to General Procedure D using propyl 4-(4-chloro-2-(4- cyanophenyl)-3-methylquinoline-7-carbonyl)piperazine-l-carboxylate as starting material (synthesized for B092)
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (d, / = 8.6 Hz, 1 H), 8.03 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.54 (d, / = 8.1 Hz, 2 H), 7.48 (d, / = 8.0 Hz, 2 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.84 (s, 2 H), 3.67- 3.26 (m, 8 H), 2.52 (s, 3 H), 1.74 (br s, 2 H), 1.65-1.37 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-(N,iV-dimethylsulfamoyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B094)
Figure imgf000226_0001
Compound B094 was synthesized according to General Procedure I, K and CI using 4-acetyl-N,N- dimethyl-benzenesulfonamide (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.53 (d, / = 8.1 Hz, 2 H), 8.49 (s, 1 H), 8.32 (d, / = 8.3 Hz, 1 H), 8.16 (s, 1 H), 7.92 (d, / = 7.9 Hz, 2 H), 7.77 (d, / = 8.4 Hz, 1 H), 4.36-3.69 (m, 6 H), 3.41-3.10 (m, 3 H), 2.71 (s, 6 H), 1.66-1.47 (m, 2 H), 1.15 (s, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 559.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(4-(4-methylpiperazin-l-yl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxyla
Figure imgf000226_0002
Compound B095 was synthesized according to General Procedure I, K and CI using 4-(4- methylpiperazino)acetophenone (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.27 (s, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 8.17 (d, / = 8.9 Hz, 2 H), 8.01 (s, 1 H), 7.63 (d, / = 7.5 Hz, 1 H), 7.05 (d, / = 8.8 Hz, 2 H), 4.37-3.63 (m, 6H), 3.21-3.09 (m, 2 H), 3.05 (s, 9 H), 2.24 (s, 3 H), 1.67-1.52 (m, 2 H), 1.13 (d, / = 5.9 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 550.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(2-(3-(4H-l,2,4-triazol-4-yl)phenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B096)
Figure imgf000226_0003
Compound B096 was synthesized according to General Procedure I, K and CI using l -[3-(4H-l,2,4- triazol-4-yl)phenyl]ethan-l-one (General Procedure I) and (R)-n-propyl 2-methylpiperazine-l- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 9.16 (s, 2 H), 8.59 (s, 1 H), 8.53 (s, 1 H), 8.41 (d, / = 7.9 Hz, 1 H), 8.31 (d, / = 8.6 Hz, 1 H), 8.16 (s, 1 H), 7.84 (d, / = 9.1 Hz, 1 H), 7.79-7.69 (m, 2 H), 4.40-3.65 (m, 6 H), 3.37-3.08 (m, 3 H), 1.67-1.53 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 519.1 [M + H+] with a purity of >99%. Propyl 4-(2-(3-(aminomethyl)-4-methoxyphenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B097)
Figure imgf000227_0001
Compound B097 was synthesized according to General Procedure L and then General Procedure D using propyl 4-(2,4-dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-cyano-4- methoxyphenyl boronic acid as starting materials (General Procedure L).
!H NMR (400 MHz, DMSO-<¾ δ 8.44 (s, 1 H), 8.33 (s, 1 H), 8.23 (dd, / = 13.1 , 5.3 Hz, 2 H), 8.10 (s, 1 H), 7.75-7.65 (m, 1 H), 7.13 (d, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.89 (s, 3 H), 3.78 (s, 2 H), 3.73-3.37 (m, 10 H), 1.59 (dd, / = 13.7, 7.1 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 497.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(4-((dimethylamino)methyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylat
Figure imgf000227_0002
Compound B098 was synthesized according to General Procedure I, K and CI using l -[4- (dimethylaminomethyl)phenyl]ethan-l -one (General Procedure I) and (R)-n-propyl 2- methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.37 (s, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.24 (d, / = 8.2 Hz, 2 H), 8.10 (s, 1 H), 7.71 (d, / = 8.4 Hz, 1 H), 7.48 (d, / = 8.2 Hz, 2 H), 4.40-3.62 (m, 6 H), 3.49 (s, 2 H), 3.34-3.13 (m, 3 H), 2.21 (s, 6 H), 1.72-1.51 (m, 2 H), 1.14 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.2 [M + H+] with a purity of >99%.
4-(4-chloro-2-(4-methoxy-3-methylphenyl)quinoline-7-carbonyl)piperazine-l-
Figure imgf000227_0003
Compound B099 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1-carboxylate and 4-methoxy-3-methylphenyl boronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.42 (s, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.21-8.11 (m, 2 H), 8.09 (s, 1 H), 7.72-7.66 (m, 1 H), 7.12 (d, / = 9.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.89 (s, 3 H), 3.73-3.37 (m, 8 H), 2.27 (s, 3 H), 1.59 (dd, / = 13.8, 6.7 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 482.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-(hydroxymethyl)-4-methoxyphenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B100)
Figure imgf000228_0001
Compound B100 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-hydroxymethyl-4-methoxyphenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.39 (s, 2 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.21 (dd, / = 8.7, 2.2 Hz, 1 H), 8.11 (s, 1 H), 7.70 (dd, / = 8.5, 1.4 Hz, 1 H), 7.13 (d, / = 8.7 Hz, 1 H), 5.16 (t, / = 5.5 Hz, 1 H), 4.59 (d, / = 5.1 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.88 (s, 3 H), 3.81-3.34 (m, 8 H), 1.59 (dd, / = 13.9, 6.9 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >97%.
4-(4-chloro-2-(4-cyano-3-methylphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000228_0002
Compound B101 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-cyano-3-methylphenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.59 (s, 1 H), 8.44 (s, 1 H), 8.32 (t, / = 7.7 Hz, 2 H), 8.19 (s, 1 H), 7.98 (d, / = 8.2 Hz, 1 H), 7.85-7.77 (m, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.73-3.36 (m, 8 H), 2.62 (s, 3 H), 1.59 (dd, / = 13.8, 7.0 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 477.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-(dimethylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B102)
Figure imgf000228_0003
B102 Compound B102 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-(dimethylcarbamoyl)phenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.56 (s, 1 H), 8.40 (d, / = 7.9 Hz, 1 H), 8.36 (s, 1 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.77 (dd, / = 8.6, 1.5 Hz, 1 H), 7.64 (t, / = 7.7 Hz, 1 H), 7.57 (d, / = 7.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.35 (m, 8 H), 3.11-2.91 (m, 6 H), 1.59 (dd, / = 13.6, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-sulfam perazine-l-carboxylate (B103)
Figure imgf000229_0001
Compound B103 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 4-sulfamoylphenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.58 (s, 1 H), 8.52 (d, / = 8.5 Hz, 2 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.19 (s, 1 H), 8.01 (d, / = 8.4 Hz, 2 H), 7.80 (d, / = 8.5 Hz, 1 H), 7.48 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.85-3.34 (m, 8 H), 1.59 (dd, / = 13.9, 7.1 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 517.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-((dimethylamino)methyl)-4-methoxyphenyl)quinoline-7- carbonyl)piperazine-l-carbo
Figure imgf000229_0002
Compound B104 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and [3-(dimethylaminomethyl)-4- methoxyphenyl] boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.40 (s, 1 H), 8.34-8.18 (m, 3 H), 8.11 (s, 1 H), 7.70 (dd, / = 8.5, 1.4 Hz, 1 H), 7.16 (d, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.88 (s, 3 H), 3.72-3.37 (m, 10 H), 2.21 (s, 6 H), 1.59 (dd, / = 13.9, 7.0 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 525.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3-(morpholinomethyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B105)
Figure imgf000230_0001
Compound B105 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 -(morpholin-4-ylmethyl)phenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.47 (s, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.26 (s, 1 H), 8.21 (d, / = 7.4 Hz, 1 H), 8.16 (s, 1 H), 7.75 (d, / = 8.5 Hz, 1 H), 7.58-7.47 (m, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.82-3.35 (m, 14 H), 2.41 (s, 4 H), 1.59 (dd, / = 14.2, 7.1 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 537.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3-cyano-4-methoxyphenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B106)
Figure imgf000230_0002
Compound B106 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-cyano-4-methoxyphenyl boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.69-8.58 (m, 2 H), 8.48 (s, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.11 (s, 1 H), 7.72 (dd, / = 8.5, 1.5 Hz, 1 H), 7.43 (d, / = 8.9 Hz, 1 H), 4.34-3.69 (m, 9 H), 3.35-3.08 (m, 3 H), 1.70-1.49 (m, 2 H), 1.14 (d, / = 5.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(2-(3-(aminomethyl)-4-methoxyphenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate
Figure imgf000230_0003
Compound B107 was synthesized according to General Procedure D using compound B106 as starting material.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.34 (s, 1 H), 8.28 (d, / = 2.2 Hz, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.18 (dd, / = 8.5, 2.2 Hz, 1 H), 8.07 (s, 1 H), 7.67 (d, / = 8.5 Hz, 1 H), 7.12 (d, / = 8.6 Hz, 1 H), 4.35-3.62 (m, 11 H), 3.36-3.12 (m, 3 H), 1.70-1.53 (m, 4 H), 1.13 (s, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 511.1 [M + H+] with a purity of >98%. Propyl 4-(2-(4-(aminomethyl)-3-methylphenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B108)
Figure imgf000231_0001
Compound B108 was synthesized according to General Procedure D using compound B101 as starting material.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37-8.31 (m, 1 H), 8.29-8.22 (m, 1 H), 8.11 (d, / = 1.1 Hz, 1 H), 8.09-7.98 (m, 2 H), 7.74-7.65 (m, 1 H), 7.58-7.44 (m, 1 H), 4.06 (dt, / = 26.9, 6.4 Hz, 2 H), 3.81 (s, 2 H), 3.69-3.32 (m, 8 H), 2.43-2.34 (m, 3 H), 1.71-1.53 (m, 4 H), 0.99-0.77 (m, 3 H).
LCMS (ESI-TOF) m/z 481.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(l-methylcyclopropyl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B109)
Figure imgf000231_0002
Step 1 : Intermediate ethyl 4-chloro-2-(prop-l-en-2-yl)quinoline-7-carboxylate was synthesized under the same conditions in General Procedure L using ethyl 2,4-dichloroquinoline-7-carboxylate and prop-l-en-2-ylboronic acid as starting material.
Step 2: To a stirred suspension of trimethylsulfoxonium iodide (1.2 g, 5.44 mmol) in dimethylsulf oxide (10 mL) and tetrahydrofuran (10 mL) was added potassium tert-butoxide (610 g, 5.44 mmol) in one portion at room temperature. After 30 min at the same temperature, a solution of ethyl 4-chloro-2-(prop-l-en-2-yl)quinoline-7-carboxylate (1 g, 3.63 mmol) in tetrahydrofuran (10 mL) was added. The resulting mixture was stirred at room temperature for 18 h and then quenched with water (20 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic layer was washed with brine and dried over anhydrous sodium sulfate, filtered, then concentrated to afford crude ethyl 4-chloro-2-(l-methylcyclopropyl)quinoline-7-carboxylate (1 g, 95%) as a brown color gum.
Step 3: 4-Chloro-2-(l-methylcyclopropyl)quinoline-7-carboxylic acid was synthesized from crude ethyl 4-chloro-2-(l-methylcyclopropyl)quinoline-7-carboxylate using General Procedure K.
Step 4: Compound B109 was synthesized according to General Procedure CI using 4-chloro-2-(l- methylcyclopropyl)quinoline-7-carboxylic acid and (R)-n-propyl 2-methylpiperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.20 (d, / = 8.0 Hz, 1 H), 7.99 (s, 1 H), 7.55 (dd, / = 1.6, 8.8 Hz, 1 H), 7.54 (s, 1 H), 4.75-2.95 (m, 9 H), 1.66 (q, / = 7.2 Hz, 2 H), 1.61 (s, 3 H), 1.45-1.35 (m, 2 H), 1.35-1.05 (m, 3 H), 1.00-0.90 (m, 5 H).
LCMS (ESI-TOF) m/z 430.2 [M + H+] with a purity of >98%. Propyl 4-(4-chloro-2-(4-((dimethylamino)methyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B110)
Figure imgf000232_0001
Compound B110 was synthesized according to General Procedure I, K and CI using l -[4- (dimethylaminomethyl)phenyl]ethan-l -one (General Procedure I) and n-propyl piperazine-1- carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.25 (dd, / = 10.8, 8.5 Hz, 3 H), 8.12 (s, 1 H), 7.72 (d, / = 8.6 Hz, 1 H), 7.48 (d, / = 8.1 Hz, 2 H), 4.00 (t, / = 6.6 Hz, 2 H), 3.64-3.34 (m, 10 H), 2.21 (s, 6 H), 1.65-1.53 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 495.2 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-(3-((dimethylamino)methyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylat Bill)
Figure imgf000232_0002
Compound Bill was synthesized according to General Procedure I, K and CI using l-[3- (dimethylaminomethyl)phenyl]ethan-l -one (General Procedure I) and (R)-n-propyl 2- methylpiperazine- 1-carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.36 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.20 (s, 1 H), 8.15 (d, / = 7.7 Hz, 1 H), 8.12 (s, 1 H), 7.72 (d, / = 7.1 Hz, 1 H), 7.55-7.43 (m, 2 H), 4.40-3.65 (m, 6 H), 3.52 (s, 2 H), 3.40-3.17 (m, 3 H), 2.22 (s, 6 H), 1.69-1.52 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3-((dimethylamino)methyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B112)
Figure imgf000232_0003
Compound B112 was synthesized according to General Procedure I, K and CI using l-[3- (dimethylaminomethyl)phenyl]ethan-l -one (General Procedure I) and n-propyl piperazine-1- carboxylate (General Procedure CI) as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.20 (s, 1 H), 8.17- 8.12 (m, 2 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.55-7.41 (m, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.64-3.41 (m, 10 H), 2.22 (s, 6 H), 1.67-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 495.2 [M + H+] with a purity of >96%.
Propyl (/?)-4-(2-(4-carbamoylphenyl)-4-chloro-3-methylquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate
Figure imgf000233_0001
Compound B113 was synthesized according to General Procedure I, K and CI using 4- propanoylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1 -carboxylate (General Procedure CI) as starting materials. Compound B113 is a side product of the synthesis of compound B114.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.29 (d, / = 8.6 Hz, 1 H), 8.04 (s, 1 H), 8.02 (d, / = 8.3 Hz, 2 H), 7.74 (dd, / = 8.6, 1.5 Hz, 1 H), 7.68 (d, / = 8.3 Hz, 2 H), 4.37-3.61 (m, 6 H), 3.40-3.13 (m, 3 H), 2.51 (s, 3 H), 1.66-1.50 (m, 2 H), 1.09 (t, / = 16.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(4-cyanophenyl)-3-methylquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B114)
Figure imgf000233_0002
Compound B114 was synthesized according to General Procedure I, K and CI using 4- propanoylbenzonitrile (General Procedure I) and (R)-n-propyl 2-methylpiperazine- 1 -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.30 (d, / = 8.6 Hz, 1 H), 8.05 (s, 1 H), 7.97 (d, / = 8.3 Hz, 2 H), 7.82 (d, / = 8.3 Hz, 2 H), 7.76 (d, / = 8.6 Hz, 1 H), 4.28-3.73 (m, 6 H), 3.34-3.17 (d, / = 9.4 Hz, 3 H), 2.50 (s, 3 H), 1.59 (dd, / = 14.1 , 6.8 Hz, 2 H), 1.12 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 491.2 [M + H+] with a purity of >98%.
4-(4-chloro-2-(2-methylpyridin-4-yl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000234_0001
Compound B115 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-methylpyridin-4-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.65 (d, / = 5.2 Hz, 1 H), 8.58 (s, 1 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.21 (s, 1 H), 8.16 (s, 1 H), 8.07 (d, / = 5.1 Hz, 1 H), 7.81 (dd, / = 8.6, 1.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.76-3.36 (m, 8 H), 2.60 (d, / = 8.1 Hz, 3 H), 1.59 (dd, / = 14.0, 7.1 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 453.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(N-methylsulfamoyl)phenyl)quinoline-7-carbonyl)piperazine-l-
Figure imgf000234_0002
Compound B116 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 4-(methylsulfamoyl)phenyl boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.58 (s, 1 H), 8.54 (d, / = 7.8 Hz, 1 H), 8.32 (d, / = 8.5 Hz, 1 H), 8.19 (s, 1 H), 7.96 (d, / = 8.4 Hz, 2 H), 7.84-7.76 (m, 1 H), 7.59 (s, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.36 (m, 8 H), 2.48 (s, 3 H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 0.94-0.78 (m, 3 H).
LCMS (ESI-TOF) m/z 531.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-sulfam perazine-l-carboxylate (B117)
Figure imgf000234_0003
B117
Compound B117 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and (3-sulfamoylphenyl)boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.80 (s, 1 H), 8.56-8.47 (m, 2 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.20 (s, 1 H), 7.99 (d, / = 7.6 Hz, 1 H), 7.83-7.74 (m, 2 H), 7.48 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.81-3.35 (m, 8 H), 1.59 (dd, / = 14.1, 7.0 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 517.1 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B118)
Figure imgf000235_0001
Compound B118 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-(N- methylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.59 (d, / = 4.6 Hz, 1 H), 8.56 (s, 1 H), 8.42 (d, / = 8.5 Hz, 2 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 8.03 (d, / = 8.4 Hz, 2 H), 7.78 (dd, / = 8.6, 1.4 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.81-3.36 (m, 8 H), 2.83 (d, / = 4.5 Hz, 3 H), 1.59 (dd, / = 13.9, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(4-((methylamino)methyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylat
Figure imgf000235_0002
Step 1 : Intermediate propyl (R)-4-(4-chloro-2-(4-formylphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate was synthesized according to General Procedure L using propyl (R)- 4-(2,4-dichloroquinoline-7-carbonyl)-2-methylpiperazine- 1 -carboxylate and 4-formylphenyl boronic acid as starting materials.
Step 2: To a solution of propyl (R)-4-(4-chloro-2-(4-formylphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (242 mg, 0.504 mmol) in methanol (5 mL) was added a 2 M solution of methylamine in tetrahydrofuran (0.52 mL, 1.04 mmol, 2.1 equiv). The mixture was cooled to 0 °C and sodium borohydride (40 mg, 1.06 mmol, 2.1 equiv) was added. After 15 min, the reaction was quenched by adding water (5 mL) and ethyl acetate (100 mL). The organic layer was separated and washed twice with water (50 mL) and thrice with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography to afford compound B119 as a yellow solid upon lyophilisation (100 mg, 40%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.23 (d, / = 8.2 Hz, 2 H), 8.09 (s, 1 H), 7.70 (d, / = 8.5 Hz, 1 H), 7.50 (d, / = 8.1 Hz, 2 H), 4.40-3.77 (m, 6 H), 3.74 (s, 2 H), 3.39-3.10 (m, 3 H), 2.33 (s, 3 H), 2.00 (br s, 1 H), 1.66-1.54 (m, 2 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-methoxy-3-((methylamino)methyl)phenyl)quinoline-7- carbonyl)piperazine-l-carboxylate (B120)
Figure imgf000236_0001
In the synthesis of compound B100, a side product propyl 4-(4-chloro-2-(3-formyl-4- methoxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate was isolated. A solution of this side product (20 mg, 0.0403 mmol) in methanol (2 mL) was added a 2 M solution of methylamine in tetrahydrofuran (0.05 mL, 0.1 mmol, 2.5 equiv). Upon cooling to 0 °C, sodium borohydride (5 mg, 0.132 mmol, 3.3 equiv) was added. After 15 min, the reaction was quenched by adding water (1 mL) and ethyl acetate (50 mL). The organic layer was separated and washed twice with water (20 mL) and thrice with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography to afford compound B120 as a yellow solid upon lyophilisation (5 mg, 24%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.42 (s, 1 H), 8.30 (d, / = 5.8 Hz, 1 H), 8.25 (d, / = 8.6 Hz, 2 H), 8.10 (s, 1 H), 7.70 (d, / = 8.5 Hz, 1 H), 7.16 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.90 (s, 3 H), 3.76 (s, 2 H), 3.74-3.37 (m, 8 H), 2.36 (s, 3 H), 1.59 (d, / = 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 511.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(2-(4-acetamidophenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B121)
Figure imgf000236_0002
Compound B121 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate and 4-acetylaminophenyl boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 9.95 (s, 1 H), 8.33 (s, 1 H), 8.25 (dd, / = 8.6, 3.7 Hz, 3 H), 8.07 (s, 1 H), 7.75 (d, / = 8.7 Hz, 2 H), 7.69 (d, / = 8.5 Hz, 1 H), 4.45-3.73 (m, 6 H), 3.35-3.13 (m, 3 H), 2.09 (s, 3 H), 1.67-1.51 (m, 2 H), 1.14 (d, / = 5.9 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 509.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(2-(4-(aminomethyl)phenyl)-4-chloro-3-methylquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate
Figure imgf000236_0003
Compound B122 was synthesized according to General Procedure D using compound B114 as starting material. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.27 (d, / = 8.6 Hz, 1 H), 8.01 (s, 1 H), 7.71 (dd, / = 8.6, 1.4 Hz, 1 H), 7.54 (d, / = 8.1 Hz, 2 H), 7.48 (d, / = 8.0 Hz, 2 H), 4.40-3.66 (m, 8 H), 3.39-3.16 (m, 3 H), 2.52 (s, 3 H), 1.66-1.52 (m, 2 H), 1.12 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 495.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B123)
Figure imgf000237_0001
Compound B123 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and l-methylindazol-5-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.78 (s, 1 H), 8.57 (s, 1 H), 8.45 (dd, / = 8.9, 1.6 Hz, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.22 (s, 1 H), 8.14 (s, 1 H), 7.82 (d, / = 8.9 Hz, 1 H), 7.73 (dd, / = 8.5, 1.5 Hz, 1 H), 4.11 (s, 3 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.82-3.38 (m, 8 H), 1.59 (dd, / = 13.9, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3-((methylamino)methyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylat
Figure imgf000237_0002
Step 1 : Step 1 : Intermediate propyl (R)-4-(4-chloro-2-(3-formylphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate was synthesized according to General Procedure L using propyl (R)- 4-(2,4-dichloroquinoline-7-carbonyl)-2-methylpiperazine- 1 -carboxylate and 3-formylphenyl boronic acid as starting materials.
Step 2: To a solution of propyl (R)-4-(4-chloro-2-(3-formylphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (350 mg, 0.729 mmol) in methanol (5 mL) was added a 2 M solution of methylamine in tetrahydrofuran (0.73 mL, 1.46 mmol, 2 equiv). The mixture was cooled to 0 °C and sodium borohydride (55 mg, 1.46 mmol, 2 equiv) was added. After 15 min, the reaction was quenched by adding water (5 mL) and ethyl acetate (100 mL). The organic layer was separated and washed twice with water (50 mL) and thrice with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude material was purified by column chromatography to afford compound B124 as a yellow solid upon lyophilisation (150 mg, 42%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.23 (s, 1 H), 8.14 (d, / = 6.3 Hz, 1 H), 8.12 (s, 1 H), 7.72 (dd, / = 8.5, 1.4 Hz, 1 H), 7.55-7.43 (m, 2 H), 4.43-3.68 (m, 8 H), 3.39-3.10 (m, 3 H), 2.34 (s, 3 H), 2.06 (br s, 1 H), 1.69-1.52 (m, 2 H), 1.14 (d, / = 5.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H). LCMS (ESI-TOF) nt/z 495.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-cyclopropylquinoline-7-carbonyl)piperazine-l-carboxylate (B125)
Figure imgf000238_0001
Compound B125 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and cyclopropylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.18 (d, / = 8.5 Hz, 1 H), 7.90 (s, 1 H), 7.80 (s, 1 H), 7.63 (dd, / = 8.5, 1.3 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.76-3.34 (m, 8 H), 2.39-2.29 (m, 1 H), 1.58 (dd, / = 14.0, 7.0 Hz, 2 H), 1.17-1.05 (m, 4 H), 0.93-0.79 (m, 3 H).
LCMS (ESI-TOF) nt/z 402.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(5-((dimethylamino)methyl)thiophen-2-yl)quinoline-7- carbonyl)piperazine-l-carboxylate B126)
Figure imgf000238_0002
Compound B126 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 5-((dimethylamino)methyl)thiophen-2- ylboronic acid, pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.29 (s, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 7.97 (d, / = 1.2 Hz, 1 H), 7.90 (d, / = 3.7 Hz, 1 H), 7.66 (dd, / = 8.5, 1.5 Hz, 1 H), 7.04 (d, / = 3.7 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.66 (s, 2 H), 3.58-3.41 (m, 8 H), 2.25 (s, 6 H), 1.69-1.51 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 501.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(2-methyl-2H-indazol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B127)
Figure imgf000238_0003
Compound B127 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 2-methylindazol-5-ylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.74 (s, 1 H), 8.54 (s, 2 H), 8.33-8.24 (m, 2 H), 8.13 (s, 1 H), 7.73 (t, / = 9.3 Hz, 2 H), 4.22 (s, 3 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.38 (m, 8 H), 1.69-1.51 (m, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(lH-indazol-5- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B128)
Figure imgf000239_0001
Compound B128 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-indazole-5-boronic acid as starting materials.
¾ NMR (400 MHz, DMSO-<¾) δ 13.29 (s, 1 H), 8.78 (s, 1 H), 8.55 (s, 1 H), 8.41 (d, / = 9.0 Hz, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.24 (s, 1 H), 8.13 (s, 1 H), 7.71 (t, / = 8.5 Hz, 2 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.78-3.38 (m, 8 H), 1.67-1.47 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(lH-indazol-6- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B129)
Figure imgf000239_0002
Compound B129 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-indazole-6-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 13.38 (s, 1 H), 8.59 (s, 1 H), 8.50 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.18 (s, 2 H), 8.11 (d, / = 8.5 Hz, 1 H), 7.94 (d, / = 8.6 Hz, 1 H), 7.76 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.36 (m, 8 H), 1.68-1.54 (m, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >96%.
4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000239_0003
Compound B130 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and l-methylpyrrole-3-boronic acid, pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.14 (d, / = 8.5 Hz, 1 H), 8.07 (s, 1 H), 7.91 (d, / = 1.0 Hz, 1 H),
7.70 (s, 1 H), 7.58 (dd, / = 8.5, 1.4 Hz, 1 H), 6.82 (dt, / = 4.4, 2.6 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H),
3.71 (s, 3 H), 3.69-3.34 (m, 8 H), 1.64-1.50 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 441.1 [M + H+] with a purity of >96%.
Propyl (/?)-4-(4-chloro-2-(2-methoxypyridin-4-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B131)
Figure imgf000240_0001
Compound B131 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 2-methoxypyridin-4-ylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.48 (s, 1 H), 8.35 (d, / = 5.4 Hz, 1 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.16 (s, 1 H), 7.83 (dd, / = 5.4, 1.4 Hz, 1 H), 7.81-7.75 (m, 1 H), 7.65 (s, 1 H), 4.45-3.65 (m, 9 H), 3.36-3.09 (m, 3 H), 1.65-1.52 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 483.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(lH-pyrrolo[2,3-b]pyridin-5-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B 132
Figure imgf000240_0002
Compound B132 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and lH-pyrrolo[2,3-b]pyridin-5- ylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 11.67 (s, 1 H), 9.17 (d, / = 2.0 Hz, 1 H), 8.84 (d, / = 2.0 Hz, 1 H), 8.47 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 7.70 (d, / = 8.5 Hz, 1 H), 7.52 (d, / = 3.4 Hz, 1 H), 6.59 (d, / = 3.4 Hz, 1 H), 4.48-3.59 (m, 6 H), 3.41-3.09 (m, 3 H), 1.69-1.52 (m, 2 H), 1.15 (d, / = 6.2 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 492.1 [M + H+] with a purity of >96%. Propyl 4-(4-chloro-2-(l-methylcyclopropyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B133)
Figure imgf000241_0001
Compound B133 was synthesized according to General Procedure CI using intermediate 4-chloro-2- (l-methylcyclopropyl)quinoline-7-carboxylic acid in the synthesis of compound B109 and n-propyl piperazine-l-carboxylate as starting materials.
*H NMR (400 MHz, CDC13) δ 8.21 (d, / = 8.4 Hz, 1 H), 7.99 (s, 1 H), 7.55 (d, / = 10.8 Hz, 1 H), 7.54 (s, 1 H), 4.08 (t, / = 6.4 Hz, 2 H), 3.80-3.50 (m, 8 H), 1.67 (dd, / = 7.2 Hz, 2 H), 1.61 (s, 3 H), 1.40- 1.38 (m, 2 H), 0.97-0.92 (m, 5 H).
LCMS (ESI-TOF) nt/z 416.2 [M + H+] with a purity of >99%.
Propyl 4-(2-(4-(lH-pyrazol-l-yl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B134)
Figure imgf000241_0002
Compound B134 was synthesized according to General Procedure I, K and CI using 4' -(lH-pyrazol- l-yl)acetophenone (General Procedure I) and n-propyl piperazine-l -carboxylate (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.66 (d, / = 2.4 Hz, 1 H), 8.56 (s, 1 H), 8.49 (d, / = 8.7 Hz, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.16 (s, 1 H), 8.07 (d, / = 8.7 Hz, 2 H), 7.83 (s, 1 H), 7.76 (d, / = 8.5 Hz, 1 H), 6.62 (s, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.35 (m, 8 H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(4-(lH-pyrazol-5-yl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B135)
Figure imgf000241_0003
Compound B135 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and [4-( 1 H-pyrazol-5-yl)phenyl] boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 12.86 (br s, 1 H), 8.42 (s, 1 H), 8.35 (d, / = 8.2 Hz, 2 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.14 (d, / = 1.1 Hz, 1 H), 7.98 (d, / = 8.0 Hz, 2 H), 7.72 (dd, / = 8.5, 1.5 Hz, 2 H), 6.78 (d, / = 1.9 Hz, 1 H), 4.00 (t, / = 6.6 Hz, 2 H), 3.69-3.37 (m, 8 H), 1.68-1.51 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(4-(lH-pyrazol-4-yl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B136)
Figure imgf000242_0001
Compound B136 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-[4-(4,4,5,5-tetramethyl-
[l ,3,2]dioxaborolan-2-yl)phenyl]-lH-pyrazole as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 12.85 (s, 1 H), 8.39 (s, 1 H), 8.29 (d, / = 8.4 Hz, 2 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.23-7.90 (m, 3 H), 7.78 (d, / = 8.4 Hz, 2 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 4.00 (t, / = 6.6 Hz, 2 H), 3.65-3.42 (m, 8 H), 1.67-1.49 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >97%.
4-(4-Chloro-7-(4-(propoxycarbonyl)piperazine-l-carbonyl)quinolin-2-yl)-2-methylbenzoic acid (B137)
Figure imgf000242_0002
Compound B137 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-borono-2-methylbenzoic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 13.04 (br s, 1 H), 8.55 (s, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.26 (s, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 8.18 (s, 1 H), 7.97 (d, / = 8.2 Hz, 1 H), 7.77 (d, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.36 (m, 8 H), 2.65 (s, 3 H), 1.70-1.45 (m, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(lH-pyrazol-5- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B138)
Figure imgf000243_0001
Compound B138 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-pyrazole-3-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 13.35 (br s, 1 H), 8.34 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.08 (s, 1 H), 7.92 (br s, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.04 (br s, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.81-3.34 (m, 8 H), 1.59 (dd, / = 13.8, 6.7 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 428.1 [M + H+] with a purity of >96%.
Propyl 4-(2-(lH-benzo[d]imidazol-5-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B139)
Figure imgf000243_0002
Compound B139 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-benzimidazole-5-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 12.69 (br s, 1 H), 8.59 (s, 1 H), 8.55 (s, 1 H), 8.34 (s, 1 H), 8.26 (t, / = 8.8 Hz, 2 H), 8.14 (s, 1 H), 7.73 (t, / = 8.3 Hz, 2 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.36 (m, 8 H), 1.59 (dd, / = 13.8, 6.9 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >99%.
4-(4-chloro-2-(l-methyl-lH-indol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000243_0003
Compound B140 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and l -methylindole-5-boronic acid, pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.58 (s, 1 H), 8.51 (s, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.21 (dd, / = 8.7, 1.4 Hz, 1 H), 8.11 (s, 1 H), 7.69 (dd, / = 8.5, 1.3 Hz, 1 H), 7.61 (d, / = 8.7 Hz, 1 H), 7.43 (d, / = 3.0 Hz, 1 H), 6.59 (d, / = 3.0 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.86 (s, 3 H), 3.78-3.35 (m, 8 1.59 (dd, / = 13.9, 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 491.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(lH-indazol-4- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B141)
Figure imgf000244_0001
Compound B141 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-indazol-4-ylboronic acid, pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 13.30 (s, 1 H), 9.05 (s, 1 H), 8.58 (s, 1 H), 8.34 (s, 1 H), 8.31 (d, / =
8.5 Hz, 1 H), 8.03 (d, / = 7.3 Hz, 1 H), 7.76 (t, / = 9.6 Hz, 2 H), 7.53 (t, / = 7.8 Hz, 1 H), 3.98 (t, / =
6.6 Hz, 2 H), 3.82-3.36 (m, 8 H), 1.63-1.50 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) m/z 478.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3-(hydroxymethyl)phenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B142)
Figure imgf000244_0002
Compound B142 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-
(hydroxymethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.28 (d, / = 8.4 Hz, 1 H), 8.25 (s, 1 H), 8.14 (d, / = 6.8 Hz, 1 H), 8.12 (s, 1 H), 7.72 (d, / = 8.4 Hz, 1 H), 7.55-7.46 (m, 2 H), 5.06 (t, / = 5.7 Hz, 1 H), 4.64 (d, / = 5.6 Hz, 2 H), 4.37-3.73 (m, 6 H), 3.18 (d, / = 4.8 Hz, 3 H), 1.68-1.51 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-hydroxy henyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B143)
Figure imgf000244_0003
B143 Compound B143 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 3-hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.28 (d, / = 7.9 Hz, 2 H), 8.12 (s, 1 H), 7.76-7.66 (m, 3 H), 7.37 (t, / = 7.9 Hz, 1 H), 6.96 (dd, / = 7.7, 1.9 Hz, 1 H), 4.01 (t, / = 6.6 Hz, 2 H), 3.69-3.36 (m, 8 H), 1.68-1.55 (m, 2 H), 0.91 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 454.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(l-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B144)
Figure imgf000245_0001
Compound B144 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and l-methylindazol-5-ylboronic acid as starting materials.
*H NMR (400 MHz, CDC13) δ 8.52 (s, 1 H), 8.30 (d, / = 8.8 Hz, 2 H), 8.18 (s, 1 H), 8.11 (d, / = 2.4 Hz, 2 H), 7.64 (d, / = 8.4 Hz, 1 H), 7.54 (d, / = 8.8 Hz, 1 H), 4.71-4.26 (m, 2 H), 4.14 (s, 3 H), 4.11- 4.03 (m, 2 H), 3.97 - 3.02 (m, 5 H), 1.72-1.56 (m, 2 H), 1.23 (br d, / = 66 Hz, 3 H), 0.95 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 506.6 [M + H+] with a purity of >98%.
Propyl (5)-4-(2-(3-(aminomethyl)phenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B145)
Figure imgf000245_0002
Compound B145 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-(aminomethyl)phenylboronic acid hydrochloride salt as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.49 (s, 1 H), 8.30-8.28 (m, 2 H), 8.19-8.11 (m, 2 H), 7.75 (br s, 1 H), 7.52 (d, / = 4.8 Hz, 2 H), 4.49-3.53 (m, 8 H), 3.22-2.97 (m, 3 H), 1.63-1.54 (m, 2 H), 1.12 (br d, / = 70 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 481.3 [M + H+] with a purity of >97%. Propyl (5)-4-(2-(4-carbamoylphenyl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B146)
Figure imgf000246_0001
Compound B146 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-
(aminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.57 (s, 1 H), 8.42 (d, / = 7.6 Hz, 2 H), 8.31 (d, / = 8.0 Hz, 1 H), 8.18-8.06 (m, 4 H), 7.78 (br s, 1 H), 7.49 (s, 1 H), 4.38-3.44 (m, 7 H), 3.18-2.99 (m, 2 H), 1.63-1.54 (m, 2 H), 1.13 (br d, / = 68 Hz, 3 H), 0.89 (t, / = 8.0 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.3 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(lH-pyrazol-4- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B147)
Figure imgf000246_0002
Compound B147 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and pyrazole-4-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 13.10 (br s, 1 H), 8.38 (br s, 2 H), 8.19 (d, / = 8.5 Hz, 1 H), 8.16 (s, 1 H), 7.98 (s, 1 H), 7.63 (dd, / = 8.6, 1.3 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.67-3.35 (m, 8 H), 1.65-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 428.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(thiophen-3-yl uinoline-7-carbonyl)piperazine-l-carboxylate (B148)
Figure imgf000246_0003
Compound B148 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and thiophene-3-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.57 (d, / = 1.9 Hz, 1 H), 8.44 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.07 (s, 1 H), 7.98 (d, / = 5.0 Hz, 1 H), 7.78-7.67 (m, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.76-3.36 (m, 8 H), 1.59 (dd, / = 13.9, 6.7 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H). LCMS (ESI-TOF) nt/z 444.0 [M + H+] with a purity of >98%.
4-(2-(6-acetamidopyridin-3-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000247_0001
Compound B149 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-acetamidopyridine-5-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 10.80 (s, 1 H), 9.26 (s, 1 H), 8.71-8.64 (m, 1 H), 8.54 (s, 1 H), 8.27 (t, / = 9.0 Hz, 2 H), 8.14 (s, 1 H), 7.75 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.75-3.38 (m, 8 H), 2.15 (s, 3 H), 1.59 (dd, / = 12.1 , 5.0 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-fluoro-3-(hydroxymethyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B150)
Figure imgf000247_0002
Compound B150 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-fluoro-3 -
(hydroxymethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.50-8.44 (m, 2 H), 8.31-8.23 (m, 2 H), 8.15 (s, 1 H), 7.75 (d, / =
8.5 Hz, 1 H), 7.35 (t, / = 9.2 Hz, 1 H), 5.41 (t, / = 5.3 Hz, 1 H), 4.66 (d, / = 4.7 Hz, 2 H), 3.98 (t, / =
6.6 Hz, 2 H), 3.79-3.36 (m, 8 H), 1.59 (dd, / = 13.8, 7.0 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.1 [M + H+] with a purity of >99%.
Propyl 4-(2-(3-carbamoyl-4-fluorophenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B151)
Figure imgf000247_0003
B151 Compound B151 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 -(aminocarbonyl)-4-fluorophenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (dd, / = 7.0, 2.1 Hz, 1 H), 8.56 (s, 1 H), 8.51-8.43 (m, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.90 (s, 1 H), 7.79-7.72 (m, 2 H), 7.49 (t, / = 9.4 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.77-3.35 (m, 8 H), 1.59 (dd, / = 13.5, 6.8 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 499.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(4-fluoro-3-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B152)
Figure imgf000248_0001
Compound B152 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-fluoro-3 -
(methylcarbamoyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.57 (s, 1 H), 8.56 (s, 2 H), 8.51-8.40 (m, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.17 (s, 1 H), 7.77 (d, / = 8.3 Hz, 1 H), 7.50 (t, / = 9.2 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.77- 3.36 (m, 8 H), 2.83 (d, / = 4.5 Hz, 3 H), 1.59 (dd, / = 13.5, 6.2 Hz, 2 H), 0.89 (t, / = 7.0 Hz, 3 H).
LCMS (ESI-TOF) m/z 513.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(thiazol-4-yl) uinoline-7-carbonyl)piperazine-l-carboxylate (B153)
Figure imgf000248_0002
B153
Compound B153 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and thiazol-4-ylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 9.33 (d, / = 1.9 Hz, 1 H), 8.63 (d, / = 1.9 Hz, 1 H), 8.48 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.12 (s, 1 H), 7.77 (d, / = 8.5 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.35 (m, 8 H), 1.59 (dd, / = 14.1, 6.8 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 445.1 [M + H+] with a purity of >97%. Propyl 4-(4-chloro-2-(3-(dimethylcarbamoyl)-4-fluorophenyl)quinoline-7-carbonyl)piperazine- 1-carboxylate (B154)
Figure imgf000249_0001
Compound B154 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 -(dimethylcarbamoyl) -4- fluorophenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.58 (s, 1 H), 8.51-8.44 (m, 1 H), 8.39 (d, / = 6.2 Hz, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.17 (s, 1 H), 7.77 (d, / = 8.5 Hz, 1 H), 7.52 (t, / = 9.0 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.76-3.36 (m, 8 H), 3.06 (s, 3 H), 2.92 (s, 3 H), 1.59 (dd, / = 14.0, 7.3 Hz, 2 H), 0.89 (t, / = 7.0 Hz, 3 H).
LCMS (ESI-TOF) m/z 527.2 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(l-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B155)
Figure imgf000249_0002
Compound B155 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and l-methylindazol-5-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.79 (s, 1 H), 8.58 (s, 1 H), 8.45 (d, / = 8.8 Hz, 1 H), 8.28 (d, / = 8.8 Hz, 1 H), 8.22 (s, 1 H), 8.12 (br d, / = 15.2 Hz, 1 H), 7.82 (d, / = 8.8 Hz, 1 H), 7.72 (br s, 1 H), 4.38 (br s, 2 H), 4.11 (s, 3 H), 4.01-3.46 (m, 5 H), 3.22-2.99 (m, 2 H), 1.63-1.54 (m, 2 H), 1.13 (br d, / = 68 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) m/z 506.3 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-hydroxy-4-methylphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B156)
Figure imgf000249_0003
Compound B156 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-hydroxy-4-methylphenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 9.64 (s, 1 H), 8.32 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.07 (s, 1 H), 7.79 (s, 1 H), 7.73 (d, / = 8.5 Hz, 1 H), 7.63 (d, / = 7.9 Hz, 1 H), 7.25 (d, / = 7.8 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.36 (m, 8 H), 2.21 (s, 3 H), 1.59 (dd, / = 13.8, 7.2 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 468.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-fluoro-3-hydroxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B157)
Figure imgf000250_0001
Compound B157 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-hydroxy-4-fluorophenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 10.24 (s, 1 H), 8.39 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.09 (s, 1 H), 7.96 (d, / = 6.9 Hz, 1 H), 7.74 (d, / = 8.6 Hz, 2 H), 7.32 (dd, / = 10.8, 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.74-3.36 (m, 8 H), 1.59 (dd, / = 13.8, 7.0 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z All .1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-(l-hydroxyethyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B158)
Figure imgf000250_0002
Compound B158 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3 -(1 -hydroxy ethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.47 (s, 1 H), 8.33-8.25 (m, 2 H), 8.16 (s, 2 H), 7.75 (d, / = 8.5 Hz, 1 H), 7.55-7.47 (m, 2 H), 5.29 (d, / = 4.1 Hz, 1 H), 4.92-4.78 (m, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.81-3.37 (m, 8 H), 1.59 (dd, / = 13.6, 6.7 Hz, 2 H), 1.41 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.2 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(3-(hydroxymethyl)-4-methylphenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B159)
Figure imgf000251_0001
Compound B159 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 -(hydroxymethyl) -4- methylphenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.42 (s, 1 H), 8.34 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.14 (s, 1 H), 8.11 (d, / = 7.5 Hz, 1 H), 7.73 (d, / = 8.7 Hz, 1 H), 7.34 (d, / = 7.9 Hz, 1 H), 5.22 (t, / = 5.4 Hz, 1 H), 4.61 (d, / = 5.3 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.76-3.38 (m, 8 H), 2.34 (s, 3 H), 1.58 (dd, / = 12.1, 5.4 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(4-hydroxyphenyl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B160)
Figure imgf000251_0002
Compound B160 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 4-hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.26 (s, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 8.15 (d, / = 8.7 Hz, 2 H), 8.01 (d, / = 1.0 Hz, 1 H), 7.63 (dd, / = 8.5, 1.5 Hz, 1 H), 6.93 (d, / = 8.7 Hz, 2 H), 4.48-3.70 (m, 6 H), 3.33-2.90 (m, 3 H), 1.65-1.53 (m, 2 H), 1.20 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 468.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-(fluoromethyl)phenyl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B161)
Figure imgf000251_0003
B 61 Compound B161 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 4-(fluoromethyl)phenylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.40 (s, 1 H), 8.34 (d, / = 7.7 Hz, 2 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.10 (d, / = 1.1 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 7.60 (d, / = 6.9 Hz, 2 H), 5.52 (d, / = 47.5 Hz, 2 H), 4.43-3.74 (m, 6 H), 3.30-2.94 (m, 3 H), 1.66-1.52 (m, 2 H), 1.21 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 484.1 [M + H+] with a purity of >95%.
Propyl (5)-4-(4-chloro-2-(3-fluoro-4-hydroxyphenyl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B162)
Figure imgf000252_0001
Compound B162 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 3-fluoro-4- hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.32 (s, 1 H), 8.23 (d, / = 8.5 Hz, 1 H), 8.09 (dd, / = 12.9, 2.1 Hz, 1 H), 8.04 (d, / = 1.1 Hz, 1 H), 7.98 (dd, / = 8.5, 1.4 Hz, 1 H), 7.66 (dd, / = 8.5, 1.5 Hz, 1 H), 7.10 (t, / = 8.8 Hz, 1 H), 4.49-3.66 (m, 6 H), 3.30-2.87 (m, 3 H), 1.68-1.52 (m, 2 H), 1.20 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 486.1 [M + H+] with a purity of >96%.
Propyl 4-(2-(benzo[d][l,3]dioxol-5-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B163)
Figure imgf000252_0002
Compound B163 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 ,4-(methylenedioxy)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.32 (s, 1 H), 8.23 (d, / = 8.6 Hz, 1 H), 8.08 (d, / = 1.4 Hz, 1 H), 7.88 (d, / = 1.9 Hz, 1 H), 7.86 (s, 1 H), 7.69 (dd, / = 8.6, 1.4 Hz, 1 H), 7.06 (d, / = 8.8 Hz, 1 H), 6.11 (s, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.63-3.38 (m, 8 H), 1.69-1.53 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >99%. Propyl 4-(2-(lH-benzo[d][l,2,3]triazol-6-yl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B164)
Figure imgf000253_0001
Compound B164 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1H- 1 ,2,3-benzotriazol-5-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.88 (s, 1 H), 8.67 (s, 1 H), 8.45 (d, / = 8.3 Hz, 1 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 8.02 (d, / = 9.0 Hz, 1 H), 7.76 (d, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.79-3.37 (m, 8 H), 1.59 (dd, / = 13.7, 7.2 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 479.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3-fluoro-4-methoxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B165)
Figure imgf000253_0002
Compound B165 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 3-fluoro-4-methoxyphenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.50 (s, 1 H), 8.26 (d, / = 8.6 Hz, 1 H), 8.24-8.17 (m, 2 H), 8.12 (s, 1 H), 7.73 (dd, / = 8.6, 1.3 Hz, 1 H), 7.36 (t, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.95 (s, 3 H), 3.78-3.35 (m, 8 H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 486.1 [M + H+] with a purity of >99%.
4-(4-chloro-2-(2-methylthiazol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000253_0003
Compound B166 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 2-methylthiazol-5-yl-boronic acid pinacol ester as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.65 (s, 1 H), 8.53 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.01 (s, 1 H), 7.72 (dd, / = 8.5, 1.1 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.36 (m, 8 H), 1.59 (dd, / = 13.8, 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 459.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3-((dimethylamino)methyl)-4-fluorophenyl)quinoline-7- carbonyl)piperazine-l-carboxylate B167)
Figure imgf000254_0001
Compound B167 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-((dimethylamino)methyl)-4- fluorophenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.49 (s, 1H), 8.38 (dd, / = 7.2, 2.1 Hz, 1H), 8.31 - 8.24 (m, 2H), 8.15 (s, 1H), 7.75 (dd, / = 8.5, 1.2 Hz, 1H), 7.37 (t, / = 9.2 Hz, 1H), 3.98 (t, / = 6.6 Hz, 2H), 3.79 - 3.37 (m, 10H), 2.22 (s, 6H), 1.59 (dd, / = 13.8, 6.8 Hz, 2H), 0.89 (t, / = 7.2 Hz, 3H).
LCMS (ESI-TOF) nt/z 513.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(l-methyl-lH-pyrazol-3-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B168)
Figure imgf000254_0002
Compound B168 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and ( 1 -methyl- 1 H-yrazol-3-yl)lboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.26 (s, 1 H), 8.25 (d, / = 8.9 Hz, 1 H), 8.07 (s, 1 H), 7.88 (d, / = 2.0 Hz, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.01 (d, / = 2.1 Hz, 1 H), 4.02-3.96 (m, 5 H), 3.77-3.36 (m, 8 H), 1.59 (dd, / = 13.6, 6.9 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 442.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(lH-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B169)
Figure imgf000255_0001
Compound B169 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and lH-indazolyl-5-boronic acid as starting materials.
!H NMR (400 MHz, 80 °C, DMSO-<¾ δ 13.12 (s, 1 H), 8.76 (s, 1 H), 8.48 (s, 1 H), 8.39 (d, / = 9.5 Hz, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.23 (s, 1 H), 8.12 (s, 1 H), 7.78-7.65 (m, 2 H), 4.42-3.61 (m, 6 H), 3.43-3.11 (m, 3 H), 1.62 (dd, / = 13.8, 6.9 Hz, 2 H), 1.17 (d, / = 5.5 Hz, 3 H), 0.92 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >95%.
Propyl (5)-4-(4-chloro-2-(2-methyl-2H-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B170)
Figure imgf000255_0002
Compound B170 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 2-methylindazolyl-5-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.70 (s, 1 H), 8.49 (s, 1 H), 8.46 (s, 1 H), 8.31-8.22 (m, 2 H), 8.12 (s, 1 H), 7.81-7.61 (m, 2 H), 4.43-3.77 (m, 9 H), 3.42-3.13 (m, 3 H), 1.71-1.54 (m, 2 H), 1.17 (d, / = 5.7 Hz, 3 H), 0.92 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 506.1 [M + H+] with a purity of >95%.
Propyl (5)-4-(4-chloro-2-(3-(hydroxymethyl)-4-methoxyphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B 171)
Figure imgf000255_0003
Compound B171 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-hydroxymethyl-4- methylphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.29 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.17 (dd, / = 8.5, 2.3 Hz, 1 H), 8.07 (s, 1 H), 7.68 (d, / = 8.5 Hz, 1 H), 7.12 (d, / = 8.6 Hz, 1 H), 4.87 (s, 1 H), 4.60 (d, / = 4.7 Hz, 2 H), 4.31-3.66 (m, 9 H), 3.44-3.10 (m, 3 H), 1.64-1.52 (m, 2 H), 1.13 (d, / = 6.0 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 512.2 [M + H+] with a purity of >96%.
Propyl 4-(2-(3-(aminomethyl)-4-fluorophenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B172)
Figure imgf000256_0001
Compound B172 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-(aminomethyl)-4-fluorophenylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.50 (s, 1 H), 8.47 (d, / = 7.1 Hz, 1 H), 8.28 (d, / = 8.6 Hz, 1 H), 8.26-8.21 (m, 1 H), 8.14 (s, 1 H), 7.75 (d, / = 8.5 Hz, 1 H), 7.33 (t, / = 9.1 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.86 (s, 2 H), 3.77-3.38 (m, 8 H), 1.59 (dd, / = 14.0, 7.1 Hz, 2 H), 0.89 (t, / = 6.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 485.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl)quinoline-7- carbonyl)piperazine-l-carboxylate B173)
Figure imgf000256_0002
Compound B173 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 3-methyl-lH-pyrazolo[3,4-b]pyridine-5- boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 13.46 (s, 1 H), 9.49 (d, / = 1.8 Hz, 1 H), 9.15 (s, 1 H), 8.67 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.75 (d, / = 8.4 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.83-3.40 (m, 8 H), 2.61 (s, 3 H), 1.59 (dd, / = 13.6, 6.7 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl)quinoline-7- carbonyl)piperazine-l-carboxylate (B174)
Figure imgf000257_0001
Compound B174 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and ( 1 -methyl- 1 H-pyrazolo[3 ,4-b]pyridin-5- yl)boronic acid pinacol ester as starting materials.
!H NMR (400 MHz, DMSO-<¾ δ 9.54 (s, 1 H), 9.17 (s, 1 H), 8.65 (s, 1 H), 8.33 (s, 1 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.76 (d, / = 8.5 Hz, 1 H), 4.14 (s, 3 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.37 (m, 8 H), 1.59 (dd, / = 12.9, 5.3 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(2-methyl-lH-pyrrolo[2,3-b]pyridin-5-yl)quinoline-7-carbonyl)piperazine- 1-carboxylate (B175)
Figure imgf000257_0002
Compound B175 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 2-methyl-lH-pyrrolo[2,3-b]pyridine-5- boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 11.73 (s, 1 H), 9.09 (s, 1 H), 8.73 (s, 1 H), 8.55 (s, 1 H), 8.26 (d, / = 8.6 Hz, 1 H), 8.13 (s, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 6.30 (s, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.80-3.39 (m, 8 H), 2.44 (s, 3 H), 1.68-1.51 (m, 2 H), 0.90 (t, / = 5.9 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(3-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B176)
Figure imgf000257_0003
Compound B176 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 3-methyl-lH-indazole-5-boronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 13.46 (s, 1 H), 9.49 (d, / = 1.8 Hz, 1 H), 9.15 (s, 1 H), 8.67 (s, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.75 (d, / = 8.4 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.84-3.39 (m, 8 H), 2.61 (s, 3 H), 1.59 (dd, / = 13.6, 6.7 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(6-methylpyridin-3-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B177)
Figure imgf000258_0001
Compound B177 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 2-picoline-5-boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 9.33 (d, / = 2.1 Hz, 1 H), 8.52 (dd, / = 8.1 , 2.2 Hz, 1 H), 8.45 (s, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.12 (s, 1 H), 7.74 (d, / = 8.4 Hz, 1 H), 7.43 (d, / = 8.2 Hz, 1 H), 4.50-3.69 (m, 6 H), 3.39-3.09 (m, 3 H), 2.57 (s, 3 H), 1.72-1.47 (m, 2 H), 1.14 (d, / = 6.0 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 467.1 [M + H+] with a purity of >95%.
Propyl (/?)-4-(4-chloro-2-(6-(methoxycarbonyl)pyridin-3-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B 178)
Figure imgf000258_0002
Compound B178 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 6-(methoxycarbonyl)pyridine-5- boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 9.57 (d, / = 1.8 Hz, 1 H), 8.82 (dd, / = 8.2, 2.3 Hz, 1 H), 8.57 (s, 1 H), 8.33 (d, / = 8.5 Hz, 1 H), 8.23-8.19 (m, 1 H), 8.18 (s, 1 H), 7.79 (d, / = 8.6 Hz, 1 H), 4.39-3.53 (m, 9 H), 3.18 (dd, / = 54.4, 40.9 Hz, 3 H), 1.59 (dt, / = 14.1 , 7.0 Hz, 2 H), 1.14 (d, / = 6.1 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 511.1 [M + H+] with a purity of >94%. Propyl 4-(4-chloro-2-(l-methyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B179)
Figure imgf000259_0001
Compound B179 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 1 -methyl- lH-pyrazol-4-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.54 (s, 1 H), 8.25-8.20 (m, 2 H), 8.20 (d, / = 8.2 Hz, 1 H), 7.97 (s, 1 H), 7.65 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.93 (s, 3 H), 3.77-3.35 (m, 8 H), 1.58 (dd, / = 13.4, 6.1 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 442.1 [M + H+] with a purity of >95%.
Propyl 4-(2-(3-amino-4-fluorophenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B180)
Figure imgf000259_0002
Compound B180 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-amino-4-fluorophenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.37 (s, 1 H), 8.26 (t, / = 8.4 Hz, 3 H), 8.12 (d, / = 1.1 Hz, 1 H), 7.72 (dd, / = 8.5, 1.5 Hz, 1 H), 7.48 (d, / = 8.2 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.63-3.41 (m, 8 H), 1.65-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 495.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-ethoxy-4-hydroxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B181)
Figure imgf000259_0003
Compound B181 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-ethoxy-4-hydroxyphenylboronic acid pinacol ester as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.30 (s, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 8.06 (s, 1 H), 7.87 (d, / = 1.9 Hz, 1 H), 7.77 (dd, / = 8.3, 2.0 Hz, 1 H), 7.65 (dd, / = 8.5, 1.3 Hz, 1 H), 6.94 (d, / = 8.3 Hz, 1 H), 4.20 (q, / = 6.9 Hz, 2 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.75-3.40 (m, 8 H), 1.68-1.51 (m, 2 H), 1.39 (t, / = 7.0 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 498.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-hydroxy-3-(hydroxymethyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B 182)
Figure imgf000260_0001
Compound B182 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-(hydroxymethyl)-4- hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.2-8.18 (m, 3 H), 8.07-7.98 (m, 2 H), 7.65 (d, / = 8.5 Hz, 1 H), 6.93 (d, / = 8.4 Hz, 1 H), 4.61 (s, 2 H), 4.33-3.64 (m, 6H), 3.36-3.15 (m, 3 H), 1.68-1.52 (m, 2 H), 1.13 (d, / = 6.5 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 498.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(l-methyl-lH-benzo[d]imidazol-5-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B183)
Figure imgf000260_0002
Compound B183 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and ( 1 -methyl- 1 H-benzimidazol-5-yl)boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.66 (d, / = 0.8 Hz, 1 H), 8.59 (s, 1 H), 8.35 (dd, / = 8.7, 1.0 Hz, 1 H), 8.30 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.15 (s, 1 H), 7.75 (d, / = 8.6 Hz, 1 H), 7.72 (dd, / = 8.9, 1.1 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.91 (s, 3 H), 3.79-3.35 (m, 8 H), 1.64-1.53 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(2-hydroxy-lH-benzo[d]imidazol-5-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B184)
Figure imgf000261_0001
Compound B184 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-hydroxybenzimidazole-5-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 10.90 (d, / = 4.9 Hz, 2 H), 8.42 (s, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.09 (s, 1 H), 7.96 (d, / = 8.6 Hz, 1 H), 7.94 (s, 1 H), 7.70 (d, / = 8.5 Hz, 1 H), 7.08 (d, / = 8.1 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.72-3.36 (m, 8 H), 1.59 (dd, / = 13.7, 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 494.1 [M + H+] with a purity of >96%.
4-(2-(2-aminopyrimidin-5-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000261_0002
Compound B185 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-aminopyrimidine-5-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 9.17 (s, 2 H), 8.43 (s, 1 H), 8.23 (d, / = 8.5 Hz, 1 H), 8.07 (s, 1 H), 7.69 (d, / = 8.3 Hz, 1 H), 7.23 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.73-3.35 (m, 8 H), 1.59 (dd, / = 14.0, 6.8 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 455.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(7-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B186)
Figure imgf000261_0003
Compound B186 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and (7-methyl-lH-indazol-5-yl)boronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 13.36 (s, 1 H), 8.60 (s, 1 H), 8.53 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.22 (s, 1 H), 8.19 (s, 1 H), 8.13 (d, / = 1.0 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.82-3.34 (m, 8 H), 2.64 (s, 3 H), 1.59 (dd, / = 14.1 , 7.1 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(lH-benzo[d]imidazol-4-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B187)
Figure imgf000262_0001
Compound B187 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and lH-benzimidazol-4-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 12.85 (s, 1 H), 8.69 (br s, 1 H), 8.39 (s, 1 H), 8.29 (d, / = 8.4 Hz, 2 H), 7.84 (s, 1 H), 7.77 (d, / = 8.4 Hz, 1 H), 7.40 (t, / = 7.7 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.80- 3.37 (m, 8 H), 1.59 (dd, / = 13.8, 7.6 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(l,5-naphthyridin-3-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B188)
Figure imgf000262_0002
Compound B188 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 1 ,5-naphthyridin-2-ylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 9.91 (d, / = 2.1 Hz, 1 H), 9.25 (s, 1 H), 9.10 (dd, / = 4.1 , 1.5 Hz, 1 H), 8.75 (s, 1 H), 8.51 (d, / = 8.0 Hz, 1 H), 8.35 (d, / = 8.6 Hz, 1 H), 8.24 (s, 1 H), 7.85 (dd, / = 8.5, 4.2 Hz, 1 H), 7.80 (dd, / = 8.6, 1.5 Hz, 1 H), 4.40-3.64 (m, 6 H), 3.39-3.12 (m, 3 H), 1.59 (dt, / = 14.2, 7.0 Hz, 2 H), 1.15 (d, / = 5.9 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B 189)
Figure imgf000263_0001
Compound B189 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-(N- methylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.46 (s, 1 H), 8.41-8.32 (m, 3 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.14 (s, 1 H), 8.00 (d, / = 8.4 Hz, 2 H), 7.75 (dd, / = 8.5, 1.3 Hz, 1 H), 4.39-3.75 (m, 6 H), 3.38-3.10 (m, 3 H), 2.84 (d, / = 4.5 Hz, 3 H), 1.66-1.51 (m, 2 H), 1.14 (d, / = 5.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 509.2 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)-3- methylpiperazine- 1-carboxylate B 190)
Figure imgf000263_0002
Compound B190 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 4-(N- methylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.45 (s, 1 H), 8.39-8.31 (m, 3 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.12 (d, / = 1.1 Hz, 1 H), 8.00 (d, / = 8.4 Hz, 2 H), 7.73 (dd, / = 8.5, 1.4 Hz, 1 H), 4.49-3.73 (m, 6 H), 3.32-3.16 (m, 2 H), 3.03-2.94 (m, 1 H), 2.84 (d, / = 4.6 Hz, 3 H), 1.67-1.50 (m, 2 H), 1.21 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 509.2 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(3-fluoro-4-hydroxyphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B191)
Figure imgf000263_0003
Compound B191 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1-carboxylate and 3-fluoro-4-hydroxyphenylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 10.47 (s, 1 H), 8.43 (s, 1 H), 8.24 (d, / = 8.4 Hz, 1 H), 8.16 (d, / = 12.8 Hz, 1 H), 8.09 (s, 1 H), 8.04 (d, / = 9.1 Hz, 1 H), 7.71 (d, / = 8.6 Hz, 1 H), 7.11 (t, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.79-3.37 (m, 8 H), 1.59 (d, / = 7.1 Hz, 2 H), 0.89 (t, / = 7.0 Hz, 3 H).
LCMS (ESI-TOF) nt/z 472.1 [M + H+] with a purity of >95%.
Propyl (5)-4-(4-chloro-2-(3-fluoro-4-hydroxyphenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B192)
Figure imgf000264_0001
Compound B192 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-fluoro-4- hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 10.14 (s, 1 H), 8.33 (s, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.14- 8.07 (m, 1 H), 8.05 (s, 1 H), 7.99 (d, / = 8.5 Hz, 1 H), 7.68 (d, / = 9.9 Hz, 1 H), 7.10 (t, / = 8.8 Hz, 1 H), 4.49-3.64 (m, 6 H), 3.39-3.10 (m, 3 H), 1.66-1.49 (m, 2 H), 1.13 (d, / = 5.9 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.1 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B193)
Figure imgf000264_0002
Compound B193 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and l-methylpyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.14 (d, / = 8.6 Hz, 1 H), 7.98 (s, 1 H), 7.88 (s, 1 H), 7.64 (s, 1 H), 7.55 (d, / = 8.3 Hz, 1 H), 6.79 (d, / = 11.5 Hz, 2 H), 4.39-3.59 (m, 9 H), 3.38-3.09 (m, 3 H), 1.59 (dd, / = 14.0, 7.0 Hz, 2 H), 1.12 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 455.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B194)
Figure imgf000265_0001
Compound B194 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l-carboxylate and l-methylpyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.13 (d, / = 8.3 Hz, 1 H), 7.97 (s, 1 H), 7.86 (d, / = 1.1 Hz, 1 H), 7.64 (s, 1 H), 7.53 (dd, / = 8.4, 1.6 Hz, 1 H), 6.79 (dt, / = 4.5, 2.7 Hz, 2 H), 4.50-3.55 (m, 9H), 3.30-3.12 (m, 2 H), 3.02-2.93 (m, 1 H), 1.59 (dd, / = 14.0, 6.6 Hz, 2 H), 1.19 (d, / = 6.8 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 455.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-methyl-lH-indazol-6-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B195)
Figure imgf000265_0002
Compound B195 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 1 -methyl- lH-indazol-6-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.70 (s, 1 H), 8.64 (s, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.20 (d, / = 10.3 Hz, 1 H), 8.19 (s, 1 H), 8.13 (s, 1 H), 7.92 (d, / = 8.6 Hz, 1 H), 7.77 (d, / = 8.6 Hz, 1 H), 4.19 (s, 3 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.39 (m, 8 H), 1.71-1.52 (m, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 492.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(2-methyl-2H-indazol-6-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B196)
Figure imgf000265_0003
Compound B196 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-methyl-2H-indazol -6-ylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 2 H), 8.42 (s, 1 H), 8.28 (d, / = 8.4 Hz, 1 H), 8.16 (d, / = 0.8 Hz, 1 H), 8.09 (dd, / = 8.8, 0.8 Hz, 1 H), 7.87 (d, / = 8.8 Hz, 1 H), 7.74 (dd, / = 8.8, 0.8 Hz, 1 H), 4.23 (s, 3 H), 3.98 (t, / = 6.8 Hz, 2 H), 3.70-3.43 (m, 8 H), 1.59 (dd, / = 14.4, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >96%.
Propyl (/?)-4-(4-chloro-2-(3-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B197)
Figure imgf000266_0001
Compound B197 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-methyl-lH-indazole-5- boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 12.64 (s, 1 H), 8.68 (s, 1 H), 8.52 (s, 1 H), 8.36 (d, / = 8.9 Hz, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.10 (s, 1 H), 7.68 (d, / = 8.6 Hz, 1 H), 7.59 (d, / = 8.7 Hz, 1 H), 4.41-3.65 (m, 6 H), 3.40-3.12 (m, 3 H), 2.60 (s, 3 H), 1.60 (dt, / = 14.2, 7.3 Hz, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 506.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(3-methyl-lH-indazol-5-yl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B198)
Figure imgf000266_0002
Compound B198 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 3-methyl-lH-indazole-5- boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 12.63 (s, 1 H), 8.67 (s, 1 H), 8.51 (s, 1 H), 8.36 (d, / = 8.8 Hz, 1 H), 8.26 (d, / = 8.4 Hz, 1 H), 8.09 (s, 1 H), 7.67 (d, / = 8.5 Hz, 1 H), 7.59 (d, / = 8.9 Hz, 1 H), 4.63-3.67 (m, 6 H), 3.35-3.14 (m, 3 H), 3.04-2.94 (m, 1 H), 1.60 (dd, / = 14.1 , 6.9 Hz, 2 H), 1.21 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 506.1 [M + H+] with a purity of >99%. Propyl (/?)-4-(4-chloro-2-(3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B 199)
Figure imgf000267_0001
Compound B199 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-methyl-lH-pyrazolo[3,4- B]pyridine-5-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 13.25 (s, 1 H), 9.45 (d, / = 2.0 Hz, 1 H), 9.08 (d, / = 2.0 Hz, 1 H), 8.58 (s, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.15 (s, 1 H), 7.72 (dd, / = 8.5, 1.5 Hz, 1 H), 4.56-3.60 (m, 6 H), 3.39-3.11 (m, 3 H), 1.67-1.52 (m, 2 H), 1.15 (d, / = 5.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(3-methyl-lH-pyrazolo[3,4-b]pyridin-5-yl)quinoline-7-carbonyl)-3- methylpiperazine- 1-carboxylate B200)
Figure imgf000267_0002
Compound B200 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 3-methyl-lH-pyrazolo[3,4- B]pyridine-5-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 13.24 (s, 1 H), 9.45 (d, / = 2.1 Hz, 1 H), 9.07 (d, / = 2.1 Hz, 1 H), 8.56 (s, 1 H), 8.28 (d, / = 8.6 Hz, 1 H), 8.13 (d, / = 1.1 Hz, 1 H), 7.70 (dd, / = 8.5, 1.6 Hz, 1 H), 4.55-3.66 (m, 6 H), 3.35-3.15 (m, 2 H), 3.03-2.89 (m, 1 H), 1.68-1.50 (m, 2 H), 1.22 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3,5-difluoro-4-hydroxyphenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B201)
Figure imgf000267_0003
Compound B201 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1-carboxylate and 3,5-difluoro-4-hydroxyphenylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 10.85 (s, 1 H), 8.51 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.16-8.04 (m, 3 H), 7.73 (d, / = 8.5 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.84-3.34 (m, 8 H), 1.59 (d, / = 6.6 Hz, 2 H), 0.90 (d, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 490.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(4-hydroxy-3-methylphenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B202)
Figure imgf000268_0001
Compound B202 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 4-hydroxy-3-methylphenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 9.90 (s, 1 H), 8.36 (s, 1 H), 8.23 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 8.07 (s, 1 H), 8.02 (d, / = 8.6 Hz, 1 H), 7.73-7.64 (m, 1 H), 6.94 (d, / = 8.5 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.39 (m, 8 H), 2.24 (s, 3 H), 1.59 (dd, / = 13.6, 6.6 Hz, 2 H), 0.90 (t, / = 7.0 Hz, 3 H).
LCMS (ESI-TOF) nt/z 468.1 [M + H+] with a purity of >97%.
Propyl 4-(2-(benzofuran-5-yl)-4-chloro uinoline-7-carbonyl)piperazine-l-carboxylate (B203)
Figure imgf000268_0002
Compound B203 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and benzofuran-5-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.65 (s, 1 H), 8.55 (s, 1 H), 8.31 (dd, / = 20.6, 8.3 Hz, 2 H), 8.13 (d, / = 17.1 Hz, 2 H), 7.76 (dd, / = 16.1 , 8.7 Hz, 2 H), 7.11 (s, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.77-3.37 (m, 8 H), 1.59 (dd, / = 14.1, 6.8 Hz, 2 H), 0.89 (t, / = 6.5 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-fluoro-4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B204)
Figure imgf000269_0001
Compound B204 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-fluoro-4-(N- methylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.36 (s, 1 H), 8.32-8.22 (m, 3 H), 8.19 (s, 1 H), 7.86- 7.75 (m, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.74-3.36 (m, 8 H), 2.82 (d, / = 4.5 Hz, 3 H), 1.59 (dd, / = 13.2, 6.3 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 513.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(4-(cyclopropylcarbamoyl)-3-fluorophenyl)quinoline-7- carbonyl)piperazine-l-carbox late (B205)
Figure imgf000269_0002
Compound B205 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-fluoro-4-(N- cyclopropylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.50 (s, 1 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.28-8.21 (m, 2 H), 8.19 (s, 1 H), 7.79 (d, / = 8.5 Hz, 1 H), 7.74 (t, / = 7.8 Hz, 1 H), 3.98 (t, / = 6.4 Hz, 2 H), 3.80- 3.36 (m, 8 H), 2.93-2.81 (m, 1H), 1.59 (dd, / = 12.4, 5.4 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H), 0.81-0.65 (m, 2 H), 0.58 (s, 2 H).
LCMS (ESI-TOF) m/z 539.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(4-(ethylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B206)
Figure imgf000269_0003
Compound B206 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(N-ethylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.62 (t, / = 5.4 Hz, 1 H), 8.57 (s, 1 H), 8.42 (d, / = 8.4 Hz, 2 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 8.03 (d, / = 8.4 Hz, 2 H), 7.78 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.74-3.39 (m, 8 H), 3.37-3.32 (m, 2 H), 1.64-1.52 (m, 2 H), 1.16 (t, / = 7.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(l-methyl-lH-benzo[d]imidazol-6-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B207)
Figure imgf000270_0001
Compound B207 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -methyl- 1 H-benzoimidazole-6-boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.52 (s, 2 H), 8.30-8.21 (m, 3 H), 8.14 (d, / = 1.0 Hz, 1 H), 7.78 (d, / = 8.7 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 4.00 (t, / = 6.5 Hz, 2 H), 3.96 (s, 3 H), 3.69- 3.40 (m, 8 H), 1.60 (dd, / = 14.2, 7.0 Hz, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 492.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(2-methylbenzo[d]thiazol-6-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B208)
Figure imgf000270_0002
Compound B208 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 2-methylbenzothiazole-6-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 9.03 (d, / = 1.1 Hz, 1 H), 8.58 (s, 1 H), 8.48 (dd, / = 8.5, 1.5 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.16 (d, / = 0.7 Hz, 1 H), 8.07 (d, / = 8.6 Hz, 1 H), 7.76 (dd, / = 8.6, 1.1 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.79-3.40 (m, 8 H), 2.86 (s, 3 H), 1.59 (dd, / = 14.0, 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(lH-pyrrol-3- l)quinoline-7-carbonyl)piperazine-l-carboxylate (B209)
Figure imgf000270_0003
B209 Compound B209 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and lH-pyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 11.31 (s, 1 H), 8.15 (d, / = 8.4 Hz, 1 H), 8.14 (s, 1 H), 7.92 (s, 1 H), 7.76 (s, 1 H), 7.58 (d, / = 8.7 Hz, 1 H), 6.89 (d, / = 1.7 Hz, 1 H), 6.84 (s, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.77-3.44 (m, 8 H), 1.59 (d, / = 8.0 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z All A [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(4-(cyclopropylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B210)
Figure imgf000271_0001
Compound B210 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 4-(N- cyclopropylaminocarbonyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.59 (d, / = 4.0 Hz, 1 H), 8.57 (s, 1 H), 8.41 (d, / = 8.1 Hz, 2 H), 8.30 (d, / = 8.4 Hz, 1 H), 8.18 (s, 1 H), 8.01 (d, / = 8.1 Hz, 2 H), 7.78 (d, / = 8.6 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.84-3.37 (m, 8 H), 2.89 (d, / = 4.6 Hz, 1 H), 1.59 (dd, / = 11.8, 5.6 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H), 0.73 (d, / = 5.9 Hz, 2 H), 0.62 (d, / = 2.1 Hz, 2 H).
LCMS (ESI-TOF) m/z 521.2 [M + H+] with a purity of >96%.
4-(4-chloro-2-(3-methyl-lH-indol-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000271_0002
Compound B211 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 3-methylindole-5-lboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 10.99 (s, 1 H), 8.55 (s, 1 H), 8.51 (s, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.15 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 7.68 (d, / = 8.5 Hz, 1 H), 7.48 (d, / = 8.6 Hz, 1 H), 7.20 (s, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.37 (m, 8 H), 2.37 (s, 3 H), 1.59 (dd, / = 13.8, 6.9 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 491.1 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(2-methylbenzo[d]oxazol-6-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B212)
Figure imgf000272_0001
Compound B212 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and (2-methyl- 1 ,3 -benzoxazol-6-yl)boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.62 (s, 1 H), 8.60 (s, 1 H), 8.39 (d, / = 8.4 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.16 (s, 1 H), 7.83 (d, / = 8.4 Hz, 1 H), 7.76 (d, / = 8.5 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.83-3.41 (m, 8 H), 2.68 (s, 3 H), 1.59 (dd, / = 15.3, 8.7 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 493.1 [M + H+] with a purity of >99%.
Propyl 4-(2-(benzo[d]oxazol-5-yl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate (B213)
Figure imgf000272_0002
Compound B213 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and l,3-benzoxazole-5-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.87 (s, 1 H), 8.78 (s, 1 H), 8.64 (s, 1 H), 8.49 (d, / = 8.4 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.97 (d, / = 8.6 Hz, 1 H), 7.76 (d, / = 8.5 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.76-3.38 (m, 8 H), 1.59 (dd, / = 12.8, 5.9 Hz, 2 H), 0.89 (t, / = 6.9 Hz, 3 H).
LCMS (ESI-TOF) m/z 479.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(l,2,5-trimethyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B214)
Figure imgf000272_0003
Compound B214 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and l,2,5-trimethylpyrrole-3-boronic acid pinacol ester as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.12 (d, / = 8.4 Hz, 1 H), 7.90 (s, 1 H), 7.82 (s, 1 H), 7.54 (d, / = 8.3 Hz, 1 H), 6.40 (s, 1 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.65-3.34 (m, 8 H), 3.45 (s, 3 H), 2.71 (s, 3 H), 2.22 (s, 3 H), 1.60 (dd, / = 14.0, 7.0 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 469.2 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-(l-methyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B215)
Figure imgf000273_0001
Compound B215 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine- 1 -carboxylate and 1 -methyl- 1 H-pyrazole-4- boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.47 (s, 1 H), 8.19 (d, / = 8.5 Hz, 1 H), 8.17 (s, 1 H), 8.10 (s, 1 H), 7.94 (s, 1 H), 7.62 (dd, / = 8.5, 1.3 Hz, 1 H), 4.34-3.61 (m, 9 H), 3.38-3.09 (m, 3 H), 1.67-1.50 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 456.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(3-fluoro-4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B216)
Figure imgf000273_0002
Compound B216 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3-fluoro-4-
(methylcarbamoyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.48 (s, 1 H), 8.30 (d, / = 8.4 Hz, 1 H), 8.26-8.13 (m, 3 H), 8.08 (br s, 1 H), 7.86-7.72 (m, 2 H), 4.30-3.78 (m, 6 H), 3.39-3.09 (m, 3 H), 2.84 (d, / = 4.4 Hz, 3 H), 1.60 (dd, / = 13.9, 6.9 Hz, 2 H), 1.15 (s, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 527.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3,5-difluoro-4-hydroxyphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B217)
Figure imgf000274_0001
Compound B217 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3,5-difluoro-4- hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.39 (s, 1 H), 8.25 (d, / = 8.6 Hz, 1 H), 8.08 (s, 1 H), 8.00 (d, / = 9.6 Hz, 2 H), 7.70 (d, / = 8.8 Hz, 1 H), 4.36-3.66 (m, 6 H), 3.37-3.12 (m, 3 H), 1.68-1.45 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-(4-hydroxy-3-methylphenyl)quinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate (B218)
Figure imgf000274_0002
Compound B218 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-hydroxy-3- methylphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.25 (s, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 8.04 (d, / = 9.9 Hz, 2 H), 7.96 (dd, / = 8.4, 2.0 Hz, 1 H), 7.64 (d, / = 8.2 Hz, 1 H), 6.93 (d, / = 8.4 Hz, 1 H), 4.37-3.69 (m, 6 H), 3.40-3.14 (m, 3 H), 2.25 (s, 3 H), 1.68-1.48 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 482.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(l-methyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B219)
Figure imgf000274_0003
Compound B219 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3 -methylpiperazine- 1 -carboxylate and 1 -methyl- 1 H-pyrazole-4- boronic acid as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.46 (s, 1 H), 8.19 (d, / = 8.6 Hz, 1 H), 8.17 (s, 1 H), 8.09 (s, 1 H), 7.93 (s, 1 H), 7.60 (d, / = 8.5 Hz, 1 H), 4.45-3.70 (m, 9 H), 3.16 (ddd, / = 33.3, 16.7, 7.2 Hz, 3 H), 1.65-1.48 (m, 2 H), 1.20 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 456.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(3,5-difluoro-4-hydroxyphenyl)quinoline-7-carbonyl)-3- methylpiperazine- 1-carboxylate B220)
Figure imgf000275_0001
Compound B220 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 3,5-difluoro-4- hydroxyphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.39 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.06 (d, / = 1.0 Hz, 1 H), 8.01 (d, / = 9.9 Hz, 2 H), 7.68 (dd, / = 8.5, 1.4 Hz, 1 H), 4.49-3.72 (m, 6 H), 3.34-3.16 (m, 3 H), 1.66-1.55 (m, 2 H), 1.20 (d, / = 6.7 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 504.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(lH-pyrrolo[2,3-b]pyridin-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B221)
Figure imgf000275_0002
Compound B221 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1-carboxylate and lH-pyrrolo[2,3-b]pyridine-4-ylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 1 1.92 (s, 1 H), 8.54 (s, 1 H), 8.42 (d, / = 5.0 Hz, 1 H), 8.34 (d, / = 8.6 Hz, 1 H), 8.28 (s, 1 H), 7.85 (d, / = 5.1 Hz, 1 H), 7.82 (dd, / = 8.6, 1.4 Hz, 1 H), 7.67 (t, / = 2.8 Hz, 1 H), 7.34 (d, / = 3.2 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.78-3.37 (m, 8 H), 1.59 (dd, / = 14.4, 7.4 Hz, 2 H), 0.90 (t, / = 6.9 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(2-methyl-lH-benzo[d]imidazol-5-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B222)
Figure imgf000276_0001
Compound B222 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and (2-methyl- 1 H- 1 ,3 -benzodiazol-6- yl)boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 12.43 (d, / = 20.7 Hz, 1 H), 8.52 (s, 1 H), 8.44 (d, / = 42.3 Hz, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.22-8.08 (m, 2 H), 7.71 (d, / = 8.8 Hz, 1 H), 7.60 (dd, / = 32.9, 7.7 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.79-3.38 (m, 8 H), 2.55 (s, 3 H), 1.59 (dd, / = 14.0, 6.4 Hz, 2 H), 0.90 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) m/z 492.1 [M + H+] with a purity of >97%. l-(4-(4-chloro-2-phenylquinoline-7-carbonyl)piperazin-l-yl)pentan-l-one (B223)
Figure imgf000276_0002
Step 1 : Intermediate from General Procedure K in the synthesis of B002 was subjected to General Procedure CI with tert- butyl piperazine-1 -carboxylate as reagent to afford teri-butyl 4-(4-chloro-2- phenylquinoline-7-carbonyl)piperazine-l -carboxylate.
Step 2: The intermediate from above was dissolved in dichloromethane and trifluoroacetic acid (1 : 1) and after 10 min, the mixture was concentrated under reduced pressure. The crude material was re- dissolved in ethyl acetate and basified with solid sodium bicarbonate and minimal amount of water. The separated organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was used without further purification.
Step 3: The crude material (88.8 mg, 0.252 mmol) was dissolved in dichloromethane (3 mL) and triethylamine (53 nL, 0.38 mmol, 1.5 equiv). To the mixture was added valeryl chloride (40 nL, 0.337 mmol, 1.3 equiv) and the mixture was quenched with saturated ammonium chloride after 20 min. The organic layer was separated and the aqueous layer was extracted twice with dichloromethane. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (0-50% ethyl acetate/hexanes) to afford B223 as a white solid (44.6 mg, 41%).
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.49 (s, 1 H), 8.33 (d, / = 7.4 Hz, 2 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.16 (s, 1 H), 7.76 (d, / = 8.6 Hz, 1 H), 7.64-7.50 (m, 3 H), 3.79-3.35 (m, 8 H), 2.33 (br s, 2 H), 1.48 (br s, 2 H), 1.31 (br s, 2 H), 0.88 (br s, 3 H).
LCMS (ESI-TOF) m/z 436.1 [M + H+] with a purity of >99%. Propyl 4-(4-chloro-2-(4-((2-methoxyethoxy)methyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B224)
Figure imgf000277_0001
Compound B224 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 4-[(2- methoxyethoxy)methyl]phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.48 (s, 1 H), 8.32 (d, / = 8.3 Hz, 2 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.14 (d, / = 1.1 Hz, 1 H), 7.75 (dd, / = 8.5, 1.5 Hz, 1 H), 7.52 (d, / = 8.3 Hz, 2 H), 4.60 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.74-3.39 (m, 12 H), 3.28 (s, 3 H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 526.1 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(4-hydroxy-3-methylphenyl)quinoline-7-carbonyl)-3-methylpiperazine- 1-carboxylate (B225)
Figure imgf000277_0002
Compound B225 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 4-hydroxy-3- methylphenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.35 (s, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 8.05-7.98 (m, 2 H), 7.65 (d, / = 8.5 Hz, 1 H), 6.93 (d, / = 8.4 Hz, 1 H), 4.16-2.90 (m, 9 H), 2.24 (s, 3 H), 1.58 (dd, / = 14.0, 7.1 Hz, 2 H), 1.18 (s, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 482.1 [M + H+] with a purity of >99%. l-(4-(4-chloroquinoline-7-carbonyl)pi erazin-l-yl)pentan-l-one (B226)
Figure imgf000277_0003
Step 1 : According to General Procedure CI, commercially available 4-chloroquinoline-7-carboxylic acid was reacted with teri-butyl-piperazine-1 -carboxylate to give teri-butyl 4-(4-chloroquinoline-7- carbonyl)piperazine-l -carboxylate. Step 2: The intermediate from above was dissolved in dichloromethane and trifluoroacetic acid (1 : 1) and after 10 min, the mixture was concentrated under reduced pressure. The crude material was re- dissolved in ethyl acetate and basified with solid sodium bicarbonate and minimal amount of water. The separated organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was used without further purification.
Step 3: To a solution of the above residue (86 mg, 0.312 mmol) in dichloromethane (3 mL) was added triethylamine (70 μL·, 0.502 mmol, 1.6 equiv) and valeryl chloride (50 μL·, 0.421 mmol, 1.3 equiv). The mixture was stirred for 30 min before quenching by the addition of saturated ammonium chloride. The aqueous layer was extracted 3 times with dichloromethane and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (0-50% ethyl acetate/hexanes) to afford B226 as colorless oil (56 mg, 50%).
*H NMR (400 MHz, DMSO-<¾) δ 8.93 (d, / = 4.7 Hz, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.13 (d, / = 0.4 Hz, 1 H), 7.86 (d, / = 4.7 Hz, 1 H), 7.80 (dd, / = 8.5, 1.3 Hz, 1 H), 3.82-3.42 (m, 8 H), 2.33 (br s, 2 H), 1.54-1.38 (m, 2 H), 1.31 (br s, 2 H), 0.88 (br s, 3 H).
LCMS (ESI-TOF) m/z 360.1 [M + H+] with a purity of >98%.
Propyl 4-(2-(2-(aminomethyl)-l,5-dimethyl-lH-pyrrol-3-yl)-4-chloroquinoline-7- carbonyl)piperazine-l-carboxylate B227)
Figure imgf000278_0001
Step 1: Propyl 4-(4-chloro-2-(2-cyano-l,5-dimethyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine- 1-carboxylate was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 2-cyano-l,5-dimethylpyrrole-3-boronic acid pinacol ester as starting materials.
Step 2: Compound B227 was synthesized according to General Procedure D using the above intermediate.
*H NMR (400 MHz, DMSO-<¾ δ 8.14 (d, / = 8.5 Hz, 1 H), 7.97 (s, 1 H), 7.92 (s, 1 H), 7.58 (d, / = 8.4 Hz, 1 H), 6.48 (s, 1 H), 4.07 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.36 (m, 13 H), 2.23 (s, 3 H), 1.58 (dd, / = 12.8, 6.4 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 484.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(l-(methoxycarbonyl)-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B228)
Figure imgf000279_0001
Compound B228 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -(methoxycarbonyl)pyrrole-3-boronic acid pinacol ester as starting materials.
!H NMR (400 MHz, DMSO-<¾) δ 8.41 (s, 1 H), 8.34 (s, 1 H), 8.22 (d, / = 8.6 Hz, 1 H), 8.03 (s, 1 H), 7.68 (dd, / = 8.6, 1.3 Hz, 1 H), 7.49-7.43 (m, 1 H), 7.12-7.05 (m, 1 H), 4.08-3.94 (m, 5 H), 3.73- 3.37 (m, 8 H), 1.59 (dd, / = 14.2, 6.8 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 485.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-isopropyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B229)
Figure imgf000279_0002
Compound B229 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and l-isopropylpyrazole-4-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.63 (s, 1 H), 8.23 (d, / = 3.7 Hz, 2 H), 8.20 (d, / = 8.5 Hz, 1 H), 7.98 (s, 1 H), 7.65 (dd, / = 8.5, 1.4 Hz, 1 H), 4.58 (dt, / = 13.3, 6.6 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.82-3.35 (m, 8H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 1.49 (d, / = 6.7 Hz, 6 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 470.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-(difluoromethyl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B230)
Figure imgf000279_0003
Compound B230 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -(difluoromethyl)pyrazole-4-boronic acid pinacol ester as starting materials. !H NMR (400 MHz, DMSO-<¾ δ 9.12 (s, 1 H), 8.57 (s, 1 H), 8.38 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.05 (s, 1 H), 7.93 (t, / = 59.0 Hz, 1 H), 7.72 (dd, / = 8.5, 1.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77- 3.36 (m, 8 H), 1.59 (dd, / = 13.9, 6.9 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 478.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(l-(N,N-dimethylsulfamoyl)-lH-pyrrol-3-yl)quinoline-7- carbonyl)piperazine-l-carboxylate B231)
Figure imgf000280_0001
02NMe2
Compound B231 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -(N,N-dimethylsulf amoyl)pyrrole-3 - boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.43 (s, 1 H), 8.24 (s, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 8.04 (s, 1 H), 7.68 (dd, / = 8.5, 1.3 Hz, 1 H), 7.43-7.31 (m, 1 H), 7.15 (dd, / = 3.0, 1.4 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.75-3.38 (m, 8 H), 2.84 (s, 6 H), 1.59 (dd, / = 14.1 , 6.9 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 534.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(l-oxoisoindolin-5-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B232)
Figure imgf000280_0002
Compound B232 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and isoindolin-l-one-5 -boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.70 (s, 1 H), 8.57 (s, 1 H), 8.53 (s, 1 H), 8.44 (d, / = 8.1 Hz, 1 H), 8.31 (d, / = 8.5 Hz, 1 H), 8.18 (s, 1 H), 7.85 (d, / = 8.0 Hz, 1 H), 7.79 (d, / = 8.6 Hz, 1 H), 4.51 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.81-3.34 (m, 8 H), 1.59 (dd, / = 14.1 , 7.0 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >99%.
3-Fluoropropyl (/?)-4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B233)
Figure imgf000281_0001
Compound B228 was synthesized according to General Procedure L using 3-fluoropropyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-
(methylcarbamoyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.45 (s, 1 H), 8.41-8.32 (m, 3 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.14 (s, 1 H), 8.00 (d, / = 8.4 Hz, 2 H), 7.75 (dd, / = 8.5, 1.2 Hz, 1 H), 4.57 (t, / = 5.9 Hz, 1 H), 4.45 (t, / = 6.0 Hz, 1 H), 4.36-3.74 (m, 6 H), 3.38-3.11 (m, 3 H), 2.84 (d, / = 4.5 Hz, 3 H), 2.04-1.90 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 527.1 [M + H+] with a purity of >95%.
3-Fluoropropyl (/?)-4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B234
Figure imgf000281_0002
Compound B234 was synthesized according to General Procedure L using 3-fluoropropyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and N-methylpyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.14 (d, / = 8.5 Hz, 1 H), 7.98 (s, 1 H), 7.88 (s, 1 H), 7.64 (s, 1 H), 7.55 (d, / = 7.2 Hz, 1 H), 6.83-6.74 (m, 2 H), 4.57 (t, / = 5.9 Hz, 1 H), 4.45 (t, / = 6.0 Hz, 1 H), 4.35-3.64 (m, 9 H), 3.34-3.09 (m, 3 H), 2.06-1.87 (m, 2 H), 1.13 (d, / = 6.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 473.1 [M + H+] with a purity of >95%.
3-Fluoropropyl (/?)-4-(4-chloro-2-(3,5-difluoro-4-hydroxyphenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B235)
Figure imgf000281_0003
Compound B235 was synthesized according to General Procedure L using 3-fluoropropyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 3,5-difluoro-4- hydroxyphenylboronic acid as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 10.48 (br s, 1 H), 8.39 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.11-7.94 (m, 3 H), 7.70 (d, / = 8.5 Hz, 1 H), 4.57 (t, / = 5.9 Hz, 1 H), 4.45 (t, / = 5.9 Hz, 1 H), 4.35- 3.57 (m, 6 H), 3.42-3.09 (m, 3 H), 2.08-1.90 (m, 2 H), 1.14 (d, / = 6.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 522.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(3-methyl-4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B236)
Figure imgf000282_0001
Compound B236 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and [3-methyl-4-
(methylcarbamoyl)phenyl]boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (s, 1 H), 8.35-8.14 (m, 5 H), 7.76 (d, / = 8.5 Hz, 1 H), 7.50 (d, / = 7.9 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.78-3.37 (m, 8 H), 2.79 (d, / = 4.6 Hz, 3 H), 1.59 (d, / = 6.4 Hz, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 509.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(2-methylbenzo[d]oxazol-5-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B237)
Figure imgf000282_0002
Compound B237 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 2-methyl-l ,3-benzoxazol-5-ylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.62 (d, / = 7.1 Hz, 1 H), 8.60 (s, 1 H), 8.39 (dd, / = 8.6, 1.6 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.17 (s, 1 H), 7.84 (d, / = 8.6 Hz, 1 H), 7.78-7.71 (m, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.38 (m, 8 H), 2.67 (s, 3 H), 1.59 (dd, / = 13.6, 6.5 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 4 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(3-fluoro-4-(pyrrolidin-l-yl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B238)
Figure imgf000283_0001
Compound B238 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-fluoro-4-pyrrolidinylphenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.34 (s, 1 H), 8.25 (d, / = 8.6 Hz, 1 H), 8.10 (d, / = 1.0 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 7.64 (dd, / = 9.1, 2.2 Hz, 1 H), 7.60-7.53 (m, 1 H), 7.19 (dd, / = 13.8, 8.3 Hz, 1 H), 3.99 (t, / = 6.5 Hz, 2 H), 3.68-3.37 (m, 12 H), 1.96 (t, / = 6.5 Hz, 4 H), 1.60 (dd, / = 14.2, 7.0 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 525.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(l-ethoxyethyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B239)
Figure imgf000283_0002
Compound B239 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-(l-ethoxyethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.26 (dd, / = 8.3, 4.3 Hz, 3 H), 8.12 (d, / = 1.1 Hz, 1 H), 7.72 (d, / = 7.0 Hz, 1 H), 7.50 (d, / = 8.1 Hz, 2 H), 4.54 (q, / = 6.4 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.69-3.29 (m, 10 H), 1.60 (dd, / = 14.0, 6.8 Hz, 2 H), 1.40 (d, / = 6.4 Hz, 3 H), 1.13 (t, / = 7.0 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 510.1 [M + H+] with a purity of >94%.
Propyl (5)-4-(4-chloro-2-(3-fluoro-4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)-3- methylpiperazine- 1-carboxylate B240)
Figure imgf000283_0003
Compound B240 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 3-fluoro-4-
(methylcarbamoyl)phenylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.40-8.34 (m, 1 H), 8.31 (d, / = 8.5 Hz, 1 H), 8.29- 8.23 (m, 2 H), 8.16 (d, / = 1.0 Hz, 1 H), 7.81 (t, / = 7.8 Hz, 1 H), 7.77 (dd, / = 8.6, 1.5 Hz, 1 H), 4.37-2.88 (m, 9 H), 2.82 (d, / = 4.6 Hz, 3 H), 1.58 (dd, / = 14.1 , 6.7 Hz, 2 H), 1.19 (s, 3 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 527.1 [M + H+] with a purity of >97%.
Propyl (5)-4-(2-(benzo[d]oxazol-5-yl)-4-chloroquinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B241)
Figure imgf000284_0001
Compound B241 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and 1 ,3-benzoxazole-5-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.86 (s, 1 H), 8.78 (d, / = 1.3 Hz, 1 H), 8.63 (s, 1 H), 8.49 (dd, / = 8.7, 1.4 Hz, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.15 (s, 1 H), 7.97 (d, / = 8.7 Hz, 1 H), 7.74 (d, / = 9.8 Hz, 1 H), 4.32-3.71 (m, 6 H), 3.25-2.83 (m, 3 H), 1.59 (d, / = 7.1 Hz, 2 H), 1.19 (d, / = 2.4 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(4-((cyclopropylmethoxy)methyl)phenyl)quinoline-7-carbonyl)piperazine- 1-carboxylate (B242)
Figure imgf000284_0002
Compound B242 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-
[(cyclopropylmethoxy)methyl]phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.48 (s, 1 H), 8.32 (d, / = 8.3 Hz, 2 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.14 (d, / = 0.9 Hz, 1 H), 7.75 (dd, / = 8.5, 1.4 Hz, 1 H), 7.52 (d, / = 8.2 Hz, 2 H), 4.58 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.38 (m, 8 H), 3.34 (d, / = 6.8 Hz, 2 H), 1.59 (dd, / = 13.8, 6.8 Hz, 2 H), 1.15-1.01 (m, 1 H), 0.89 (t, / = 7.2 Hz, 3 H), 0.54-0.45 (m, 2 H), 0.24-0.14 (m, 2 H).
LCMS (ESI-TOF) nt/z 522.2 [M + H+] with a purity of >96%.
Propyl (/?)-4-(2-(benzo[d]oxazol-5-yl)-4-chloroquinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B243)
Figure imgf000285_0001
Compound B243 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 1 ,3-benzoxazole-5-boronic acid pinacol ester as starting materials.
!H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.76 (s, 1 H), 8.72 (s, 1 H), 8.52 (s, 1 H), 8.44 (dd, / = 8.6, 1.5 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.14 (s, 1 H), 7.91 (d, / = 8.7 Hz, 1 H), 7.73 (dd, / = 8.5, 1.3 Hz, 1 H), 4.41-3.59 (m,6 H), 3.22 (dd, / = 35.0, 20.1 Hz, 3 H), 1.66-1.53 (m, 2 H), 1.15 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B244)
Figure imgf000285_0002
Compound B244 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(2-hydroxy-2-propanyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.46 (s, 1 H), 8.30-8.22 (m, 3 H), 8.13 (s, 1 H), 7.74 (d, / = 8.5 Hz, 1 H), 7.66 (d, / = 8.4 Hz, 2 H), 5.15 (s, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.39 (m, 8 H), 1.59 (dd, / = 13.7, 6.8 Hz, 2 H), 1.48 (s, 6 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >98%.
Isobutyl 4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B245)
Figure imgf000285_0003
Compound B245 was synthesized according to General Procedure L using isobutyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(N-methylcarbamoyl)phenylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.63-8.57 (m, 1 H), 8.57 (s, 1 H), 8.42 (d, / = 8.4 Hz, 2 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.18 (s, 1 H), 8.02 (d, / = 8.4 Hz, 2 H), 7.78 (dd, / = 8.5, 1.1 Hz, 1 H), 3.82 (d, / = 6.5 Hz, 2 H), 3.77-3.40 (m, 8 H), 2.83 (d, / = 4.5 Hz, 3 H), 1.87 (s, 1 H), 0.89 (d, / = 6.4 Hz, 6 H).
LCMS (ESI-TOF) nt/z 509.1 [M + H+] with a purity of >98%.
Isobutyl 4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l-carboxylate (B246)
Figure imgf000286_0001
Compound B246 was synthesized according to General Procedure L using isobutyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and l -methylpyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.14 (d, / = 8.5 Hz, 1 H), 8.07 (s, 1 H), 7.91 (s, 1 H), 7.70 (s, 1 H), 7.61-7.53 (m, 1 H), 6.88-6.77 (m, 2 H), 3.81 (d, / = 6.5 Hz, 2 H), 3.71 (s, 3 H), 3.69-3.34 (m, 8 H), 1.96-1.75 (m, 1 H), 0.89 (d, / = 6.5 Hz, 6 H).
LCMS (ESI-TOF) nt/z 455.1 [M + H+] with a purity of >98%.
2-Fluoroethyl 4-(4-chloro-2-(4-(methylcarbamoyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B247)
Figure imgf000286_0002
Compound B247 was synthesized according to General Procedure L using 2-fluoroethyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(N-methylcarbamoyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.61 - 8.57 (m, 1H), 8.57 (s, 1H), 8.42 (d, / = 8.4 Hz, 2H), 8.30 (d, / = 8.5 Hz, 1H), 8.18 (s, 1H), 8.02 (d, / = 8.4 Hz, 2H), 7.78 (dd, / = 8.5, 1.3 Hz, 1H), 4.61 (d, / = 47.6 Hz, 2H), 4.37 - 4.20 (m, 2H), 3.81 - 3.40 (m, 8H), 2.83 (d, / = 4.5 Hz, 3H).
LCMS (ESI-TOF) nt/z 499.1 [M + H+] with a purity of >97%.
2-Fluoroethyl 4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B248)
Figure imgf000287_0001
Compound B248 was synthesized according to General Procedure L using 2-f uoroethyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and l-methylpyrrole-3-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.15 (d, / = 8.5 Hz, 1H), 8.07 (s, 1H), 7.92 (s, 1H), 7.71 (s, 1H), 7.58 (d, / = 8.5 Hz, 1H), 6.83 (d, / = 16.7 Hz, 2H), 4.61 (d, / = 46.7 Hz, 2H), 4.45 - 4.16 (m, 2H), 3.71 (s, 5H), 3.43 (s, 7H).
LCMS (ESI-TOF) m/z 445.1 [M + H+] with a purity of >98%.
Propyl 4-(2-(l-(2-amino-2-oxoethyl)-lH-pyrazol-4-yl)-4-chloroquinoline-7-carbonyl)piperazine- 1-carboxylate (B249)
Figure imgf000287_0002
Compound B249 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -(2-amino-2-oxoethyl)- 1 H-pyrazol-4- ylboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.54 (s, 1 H), 8.26 (s, 1 H), 8.23 (s, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 7.99 (s, 1 H), 7.70-7.62 (m, 1 H), 7.59 (s, 1 H), 7.32 (s, 1 H), 4.87 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.33 (m, 8 H), 1.59 (dd, / = 14.0, 7.0 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 485.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(5-(trifluoromethyl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B250)
Figure imgf000287_0003
Compound B250 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 5-trifluoromethyl-lH-pyrazol-4-ylboronic acid as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 13.99 (br s, 1 H), 8.82 (s, 1 H), 8.25 (d, / = 8.5 Hz, 1 H), 8.20 (s, 1 H), 7.97 (s, 1 H), 7.73 (d, / = 8.7 Hz, 1 H), 3.98 (t, / = 6.5 Hz, 2 H), 3.81-3.34 (m, 8 H), 1.58 (dd, / = 12.0, 5.6 Hz, 2 H), 0.89 (t, / = 6.9 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(lH-pyrrol-3-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B251)
Figure imgf000288_0001
Compound B251 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and lH-pyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 1 1.10 (s, 1 H), 8.14 (d, / = 8.4 Hz, 1 H), 8.03 (s, 1 H), 7.90 (s, 1 H), 7.68 (s, 1 H), 7.55 (dd, / = 8.5, 1.5 Hz, 1 H), 6.84 (d, / = 13.5 Hz, 2 H), 4.41-3.69 (m, 6 H), 3.32-3.11 (m, 3 H), 1.67-1.47 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 441.0 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(l-(methylsulfonyl)-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B252)
Figure imgf000288_0002
Compound B252 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -(methylsulfonyl)pyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.26 (s, 1 H), 8.21 (t, / = 8.8 Hz, 1 H), 8.17 (t, / = 1.8 Hz, 1 H), 8.02 (d, / = 1.1 Hz, 1 H), 7.67 (dd, / = 8.5, 1.5 Hz, 1 H), 7.40-7.35 (m, 1 H), 7.14 (dd, / = 3.2, 1.6 Hz, 1 H), 3.99 (t, / = 6.6 Hz, 2 H), 3.50 (d, / = 26.1 Hz, 11 H), 1.65-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 505.1 [M + H+] with a purity of >99%.
Propyl 4-(2-(4-carbamoyl-3,5-difluorophenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B253)
Figure imgf000289_0001
Compound B253 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 ,5 -difluoro-4-(carbamoyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.52 (s, 1 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.18 (s, 1 H), 8.07 (d, / = 8.8 Hz, 2 H), 7.98 (s, 1 H), 7.78 (dd, / = 8.5, 1.3 Hz, 1 H), 7.65 (s, 1 H), 4.00 (t, / = 6.5 Hz, 2 H), 3.66-3.39 (m, 8 H), 1.65-1.51 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 517.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(lH-pyrrol-3-yl)quinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B254)
Figure imgf000289_0002
Compound B254 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and lH-pyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 11.14 (br s, 1 H), 8.14 (d, / = 8.4 Hz, 1 H), 8.04 (s, 1 H), 7.88 (s, 1 H), 7.69 (s, 1 H), 7.53 (dd, / = 8.5, 1.5 Hz, 1 H), 6.89-6.80 (m, 2 H), 4.47-3.73 (m, 6 H), 3.35-2.88 (m, 3 H), 1.65-1.53 (m, 2 H), 1.19 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 441.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-2-(4-(hydroxymethyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B255)
Figure imgf000289_0003
Compound B255 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-(hydroxymethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.26 (dd, / = 8.4, 5.1 Hz, 3 H), 8.12 (d, / = 1.1 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 7.51 (d, / = 8.2 Hz, 2 H), 5.05 (br s, 1 H), 4.61 (s, 2 H), 4.00 (t, / = 6.6 Hz, 2 H), 3.72-3.39 (m, 8 H), 1.70-1.52 (m, 2 H), 0.90 (t, / = 7.4 Hz, 3 H). LCMS (ESI-TOF) nt/z 468.1 [M + H+] with a purity of >99%.
4-(4-chloro-2-(4-(ethoxymethyl)phenyl)quinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000290_0001
Compound B256 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l -carboxylate and 4-(ethoxymethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 7.9 Hz, 3 H), 8.12 (s, 1 H), 7.72 (d, / = 8.5 Hz, 1 H), 7.50 (d, / = 8.1 Hz, 2 H), 4.56 (s, 2 H), 4.00 (t, / = 6.5 Hz, 2 H), 3.65-3.42 (m, 10 H), 1.72-1.51 (m, 2 H), 1.20 (t, / = 7.0 Hz, 3 H), 0.90 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 496.1 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(3-fluoro-4-(hydroxymethyl)phenyl)quinoline-7-carbonyl)piperazine-l- carboxylate (B257)
Figure imgf000290_0002
Compound B257 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3-fluoro-4-
(hydroxymethyl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.53 (s, 1 H), 8.29 (d, / = 8.6 Hz, 1 H), 8.20 (dd, / = 8.0, 1.5 Hz, 1 H), 8.16 (d, / = 1.1 Hz, 1 H), 8.12 (dd, / = 11.7, 1.5 Hz, 1 H), 7.76 (dd, / = 8.5, 1.6 Hz, 1 H), 7.67 (t, / = 7.9 Hz, 1 H), 5.42 (br s, 1 H), 4.65 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.48 (m, 8 H), 1.59 (dd, / = 14.0, 7.0 Hz, 2 H), 0.89 (t, / = 7.1 Hz, 3 H).
LCMS (ESI-TOF) nt/z 486.1 [M + H+] with a purity of >99%.
Propyl 4-(2-(4-(aminomethyl)-3,5-difluorophenyl)-4-chloroquinoline-7-carbonyl)piperazine-l- carboxylate (B258)
Figure imgf000291_0001
Compound B258 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 3 ,5 -difluoro-4-
(aminomethyl)phenylboronic acid as starting materials.
LCMS (ESI-TOF) nt/z 503.1 [M + H+] with a purity of >94%.
( ?)-4-(4-chloro-2-cyclopropylquinoline-7-carbonyl)-2-methylpiperazine-l-
Figure imgf000291_0002
Compound B259 was synthesized according to General Procedure L using 2-fluoroethyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and cyclopropyl boronic acid as starting materials.
*H NMR (600 MHz, DMSO-<¾ δ 8.19 (d, / = 8.5 Hz, 1 H), 7.88 (d, / = 29.3 Hz, 1 H), 7.80 (s, 1 H), 7.63 (d, / = 14.7 Hz, 1 H), 4.64 (t, / = 3.8 Hz, 1 H), 4.56 (t, / = 3.8 Hz, 1 H), 4.46-4.17 (m, 3 H), 3.99 (d, / = 124.3 Hz, 1 H), 3.63 (d, / = 101.9 Hz, 1 H), 3.50-3.36 (m, 1 H), 3.24-2.90 (m, 3 H), 2.36-2.26 (m, 1 H), 1.26-0.94 (m, 7 H).
LCMS (ESI-TOF) nt/z 420.1 [M + H+] with a purity of >98%.
2-Fluoroethyl (5)-4-(4-chloro-2-cyclopropylquinoline-7-carbonyl)-3-methylpiperazine-l- carboxylate (B260)
Figure imgf000291_0003
Compound B260 was synthesized according to General Procedure L using 2-fluoroethyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-3-methylpiperazine-l -carboxylate and cyclopropyl boronic acid as starting materials.
*H NMR (600 MHz, DMSO-<¾ δ 8.19 (d, / = 8.5 Hz, 1 H), 7.89 (d, / = 1.1 Hz, 1 H), 7.81 (s, 1 H), 7.63 (dd, / = 8.5, 1.4 Hz, 1 H), 4.89-3.61 (m, 8 H), 3.10 (dd, / = 112.3, 62.2 Hz, 3 H), 2.38-2.30 (m, 1 H), 1.23-1.01 (m, 7 H).
LCMS (ESI-TOF) nt/z 420.1 [M + H+] with a purity of >99%. 4-(4-chloro-2-(l-methyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l-
Figure imgf000292_0001
Compound B261 was synthesized according to General Procedure L using 2-fluoroethyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and 1 -methyl- lH-pyrazole-4-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.54 (s, 1 H), 8.25-8.16 (m, 3 H), 7.98 (d, / = 1.1 Hz, 1 H), 7.66 (dd, / = 8.5, 1.5 Hz, 1 H), 4.61 (d, / = 48.0 Hz, 2 H), 4.37-4.21 (m, 2 H), 3.93 (s, 3 H), 3.77-3.40 (m, 8 H).
LCMS (ESI-TOF) m/z 446.1 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-3-fluoro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B262)
Figure imgf000292_0002
Propyl 4-(2,4-dichloro-3-fluoroquinoline-7-carbonyl)piperazine-l-carboxylate was synthesized using General Procedure 2 for synthesis of quinolines using 2-fluoro-propanedioic acid instead of malonic acid as starting material. Compound B262 was then synthesized according to General Procedure L using propyl 4-(2,4-dichloro-3-fluoroquinoline-7-carbonyl)piperazine-l-carboxylate and N- methylpyrrole-3-boronic acid pinacol ester as starting materials.
*H NMR (600 MHz, DMSO-<¾ δ 8.13 (d, / = 8.5 Hz, 1 H), 7.97 (d, / = 1.3 Hz, 1 H), 7.69-7.62 (m, 2 H), 6.92 (t, / = 2.3 Hz, 1 H), 6.86 (d, / = 1.3 Hz, 1 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.79-3.33 (m, 11 H), 1.64-1.51 (m, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 459.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(l-fluorocyclopropyl)quinoline-7-carbonyl)piperazine-l-carboxylate (B263)
Figure imgf000292_0003
Compound B263 was synthesized using General Procedure I, K and C using 1 -fluorocyclopropyl methyl ketone (General Procedure I) and propyl piperazine-l-carboxylate (General Procedure C) as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.28 (d, / = 8.5 Hz, 1 H), 7.98 (s, 2 H), 7.74 (dd, / = 8.5, 1.4 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.72-3.45 (m, 8 H), 1.75-1.50 (m, 6 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 420.1 [M + H+] with a purity of >97%.
4-(2-(4-(azidomethyl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine-l-carboxylate
Figure imgf000293_0001
Compound B264 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(azidomethyl)benzeneboronic acid pinacol ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.51 (s, 1 H), 8.37 (d, / = 8.3 Hz, 2 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.15 (d, / = 1.2 Hz, 1 H), 7.76 (dd, / = 8.5, 1.6 Hz, 1 H), 7.58 (d, / = 8.3 Hz, 2 H), 4.58 (s, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.75- 3.34 (m, 8 H), 1.59 (dd, / = 14.0, 7.1 Hz, 2 H), 0.89 (t, / = 7.3 Hz, 3 H).
LCMS (ESI-TOF) nt/z 493.1 [M + H+] with a purity of >97%.
Propyl (/?)-4-(4-chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B265)
Figure imgf000293_0002
Compound B265 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-(2~hydroxypropan~2- yl)phenylboronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 2 H), 8.09 (s, 1 H), 7.71 (d, / = 8.5 Hz, 1 H), 7.65 (d, / = 8.4 Hz, 2 H), 4.88 (s, 1 H), 4.30-3.73 (m, 6 H), 3.34-3.11 (m, 3 H), 1.66-1.54 (m, 2 H), 1.50 (s, 6 H), 1.14 (d, / = 6.3 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 510.2 [M + H+] with a purity of >97%.
2-Fluoroethyl 4-(4-chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)piperazine- 1-carboxylate (B266)
Figure imgf000294_0001
Compound B266 was synthesized according to General Procedure L using 2-fluoroethyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 4-(2-hydroxypropan-2-yl)phenylboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.46 (s, 1 H), 8.30-8.22 (m, 3 H), 8.14 (d, / = 1.2 Hz, 1 H), 7.74 (dd, / = 8.5, 1.5 Hz, 1 H), 7.66 (d, / = 8.5 Hz, 2 H), 5.14 (s, 1 H), 4.61 (d, / = 47.8 Hz, 2 H), 4.37- 4.19 (m, 2 H), 3.79-3.35 (m, 8 H), 1.48 (s, 6 H).
LCMS (ESI-TOF) m/z 500.1 [M + H+] with a purity of >99%.
2-Fluoroethyl (/?)-4-(4-chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B267)
Figure imgf000294_0002
Compound B267 was synthesized according to General Procedure L using 2-fluoroethyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-(2-hydroxypropan-2- yl)phenyi boron ie acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.36 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.22 (d, / = 8.4 Hz, 2 H), 8.10 (s, 1 H), 7.71 (dd, / = 8.5, 1.3 Hz, 1 H), 7.65 (d, / = 8.4 Hz, 2 H), 4.88 (s, 1 H), 4.59 (dt, / = 47.8, 4.2 Hz, 2 H), 4.34-3.63 (m, 8 H), 3.36-3.12 (m, 3 H), 1.50 (s, 6 H), 1.15 (d, / = 6.3 Hz, 3 H).
LCMS (ESI-TOF) m/z 514.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)-2-methylpiperazine-l- carboxylate (B268)
Figure imgf000294_0003
Compound B268 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 1 -methylpyrroie~3-boromc acid pinacol ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.14 (d, / = 8.4 Hz, 1 H), 7.99 (s, 1 H), 7.88 (d, / = 1.1 Hz, 1 H), 7.65 (s, 1 H), 7.55 (dd, / = 8.5, 1.5 Hz, 1 H), 6.79 (dt, / = 4.4, 2.6 Hz, 2 H), 4.33-3.74 (m, 6 H), 3.70 (s, 3 H), 3.35-3.12 (m, 3 H), 1.64-1.53 (m, 2 H), 1.12 (d, / = 6.1 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H). LCMS (ESI-TOF) nt/z 455.2 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(3-fluoro-4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B269)
Figure imgf000295_0001
Compound B269 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and (3-tluoro~4-(2-hydroxypropan- 2-yI)phenyl)boronic acid pinacol ester as starting materials.
!H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.42 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.11 (s, 1 H), 8.11- 8.07 (m, 1 H), 8.02 (dd, / = 13.5, 1.7 Hz, 1 H), 7.82 (t, / = 8.4 Hz, 1 H), 7.73 (dd, / = 8.5, 1.5 Hz, 1 H), 5.14 (s, 1 H), 4.31-3.75 (m, 6 H), 3.38-3.12 (m, 3 H), 1.61 (dd, / = 14.1 , 7.3 Hz, 2 H), 1.56 (s, 6 H), 1.14 (d, / = 5.9 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 528.2 [M + H+] with a purity of >95%.
Propyl (5)-4-(4-chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B270)
Figure imgf000295_0002
Compound B270 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 4-(2-hydroxypropan-2- y phenyi boron ie acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.36 (s, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.22 (d, / = 8.5 Hz, 2 H), 8.09 (d, / = 1.1 Hz, 1 H), 7.71 (dd, / = 8.5, 1.5 Hz, 1 H), 7.65 (d, / = 8.5 Hz, 2 H), 4.87 (s, 1 H), 4.31-3.76 (m, 6 H), 3.39-3.10 (m, 3 H), 1.66-1.54 (m, 2 H), 1.50 (s, 6 H), 1.14 (d, / = 6.7 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 510.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(l-(oxetan-3-yl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B271)
Figure imgf000296_0001
Compound B271 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and l-(3-oxeta»yl)-lH-pyrazole-4-boronic acid pinaeoi ester as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.78 (s, 1 H), 8.39 (s, 1 H), 8.27 (s, 1 H), 8.21 (d, / = 8.5 Hz, 1 H), 7.99 (d, / = 0.8 Hz, 1 H), 7.67 (dd, / = 8.6, 1.4 Hz, 1 H), 5.67 (p, / = 7.4 Hz, 1 H), 4.96 (p, / = 6.8 Hz, 4 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.77-3.35 (m, 8 H), 1.64-1.52 (m, 2 H), 0.89 (t, / = 7.5 Hz, 3 H).
LCMS (ESI-TOF) m/z 484.1 [M + H+] with a purity of >99%.
Propyl (/?)-4-(4-chloro-2-(l-(oxetan-3-yl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B272
Figure imgf000296_0002
Compound B272 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 1 -(3-oxetanyl)- 1 H-pyrazole-4- boronic acid pinaeoi ester as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.71 (s, 1 H), 8.33 (s, 1 H), 8.21 (d, / = 8.4 Hz, 1 H), 8.18 (s, 1 H), 7.96 (d, / = 1.0 Hz, 1 H), 7.64 (dd, / = 8.5, 1.4 Hz, 1 H), 5.64 (p, / = 6.7 Hz, 1 H), 4.97 (p, / = 6.7 Hz, 4 H), 4.29-3.73 (m, 6 H), 3.34-3.09 (m, 3 H), 1.67-1.52 (m, 2 H), 1.13 (d, / = 6.0 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 498.2 [M + H+] with a purity of >98%.
Propyl (5)-4-(4-chloro-2-(l-(oxetan-3-yl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B273
Figure imgf000296_0003
Compound B273 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and I-(3-oxetanyl)-lH-pyi"azo]e-4- boronic acid pina oi ester as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.71 (s, 1 H), 8.33 (s, 1 H), 8.20 (t, / = 7.6 Hz, 1 H), 8.17 (s, 1 H), 7.96 (s, 1 H), 7.64 (dd, / = 8.5, 1.3 Hz, 1 H), 5.71-5.54 (m, 1 H), 4.97 (p, / = 6.7 Hz, 4 H), 4.33-3.59 (m, 6 H), 3.36-3.07 (m, 3 H), 1.66-1.52 (m, 2 H), 1.13 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 498.2 [M + H+] with a purity of >97%.
Propyl 4-(4-chloro-2-(l-(2,2,2-trifluoroethyl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine- 1-carboxylate (B274)
Figure imgf000297_0001
Compound B274 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine-l-carboxylate and (l-(2,2,2-ii'iilQoroethyl)-l H-pyrazol-4- yljboronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.70 (s, 1 H), 8.41 (s, 1 H), 8.30 (s, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 8.01 (s, 1 H), 7.68 (dd, / = 8.5, 1.4 Hz, 1 H), 5.27 (q, / = 9.3 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.75- 3.37 (m, 8 H), 1.59 (dd, / = 13.2, 6.1 Hz, 2 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 510.1 [M + H+] with a purity of >98%.
Propyl (/?)-4-(4-chloro-2-(l-(2,2,2-trifluoroethyl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B275)
Figure imgf000297_0002
Compound B275 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and (1 -(2,2,2-trifl.uoroethyl)- J.H- pyf azol-4-yl)boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.66 (s, 1 H), 8.35 (s, 1 H), 8.22 (d, / = 8.4 Hz, 1 H), 8.20 (s, 1 H), 7.98 (d, / = 1.2 Hz, 1 H), 7.66 (dd, / = 8.3, 1.1 Hz, 1 H), 5.19 (q, / = 9.1 Hz, 2 H), 4.33-3.69 (m, 6 H), 3.32-3.09 (m, 3 H), 1.67-1.52 (m, 2 H), 1.13 (d, / = 6.6 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 524.1 [M + H+] with a purity of >96%.
Propyl (5)-4-(4-chloro-2-(l-(2,2,2-trifluoroethyl)-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B276)
Figure imgf000298_0001
Compound B276 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and (l -(2,2,2-trifSuoroethyl)- lH- pyf azol-4-yl)boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.65 (s, 1 H), 8.35 (s, 1 H), 8.22 (d, / = 8.5 Hz, 1 H), 8.19 (s, 1 H), 7.98 (d, / = 0.8 Hz, 1 H), 7.65 (dd, / = 8.2, 1.3 Hz, 1 H), 5.28-5.08 (m, 2 H), 4.30-3.73 (m, 6 H), 3.36-3.09 (m, 3 H), 1.66-1.47 (m, 2 H), 1.13 (d, / = 6.2 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 524.1 [M + H+] with a purity of >95%.
Propyl 4-(4-chloro-2-(l-cyclopentyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)piperazine-l- carboxylate (B277)
Figure imgf000298_0002
Compound B277 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and 1 -cyelopentyl- 1 H-pyrazole-4-boronic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.63 (s, 1 H), 8.24 (d, / = 1.3 Hz, 2 H), 8.20 (d, / = 8.4 Hz, 1 H), 7.98 (s, 1 H), 7.65 (d, / = 8.6 Hz, 1 H), 4.84-4.68 (m, 1 H), 3.98 (t, / = 6.7 Hz, 2 H), 3.77-3.37 (m, 8 H), 2.21-2.08 (m, 2 H), 2.05-1.93 (m, 2 H), 1.89-1.77 (m, 2 H), 1.72-1.63 (m, 2 H), 1.63-1.49 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 496.2 [M + H+] with a purity of >96%.
Propyl (/?)-4-(4-chloro-2-(l-cyclopentyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B278
Figure imgf000298_0003
Compound B278 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and l-cyciopentyl-lH-pyrazole-4- boronic acid as starting materials. *H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.55 (s, 1 H), 8.23-8.17 (m, 2 H), 8.15 (s, 1 H), 7.95 (d, / = 1.0 Hz, 1 H), 7.62 (dd, / = 8.8, 1.2 Hz, 1 H), 4.82-4.70 (m, 1 H), 4.33-3.74 (m, 6 H), 3.36-3.15 (m, 3 H), 2.23-2.09 (m, 2 H), 2.08-1.93 (m, 2 H), 1.89-1.75 (m, 2 H), 1.75-1.65 (m, 2 H), 1.59 (dd, / = 14.2, 7.4 Hz, 2 H), 1.12 (d, / = 4.5 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 510.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(4-chloro-2-(l-cyclopentyl-lH-pyrazol-4-yl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B279
Figure imgf000299_0001
Compound B279 was synthesized according to General Procedure L using propyl (5)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and 1 -cyclopeniyl-lH-pyrazole-4- boronic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.55 (s, 1 H), 8.23-8.17 (m, 2 H), 8.15 (s, 1 H), 7.95 (d, / = 1.0 Hz, 1 H), 7.62 (dd, / = 8.8, 1.2 Hz, 1 H), 4.82-4.70 (m, 1 H), 4.33-3.74 (m, 6 H), 3.36-3.15 (m, 3 H), 2.23-2.09 (m, 2 H), 2.08-1.93 (m, 2 H), 1.89-1.75 (m, 2 H), 1.75-1.65 (m, 2 H), 1.59 (dd, / = 14.2, 7.4 Hz, 2 H), 1.12 (d, / = 4.5 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 510.2 [M + H+] with a purity of >99%.
Propyl (5)-4-(2-(3-(aminomethyl)-4-methoxyphenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B280)
Figure imgf000299_0002
Compound B280 was synthesized according to General Procedure L and then General Procedure D using propyl (5)-4-(2,4-dichloroquinoline-7-carbonyl)-2-methylpiperazine- 1-carboxylate and 3- cyano-4-methoxyphenyl boronic acid as starting materials (General Procedure L).
¾ NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.34 (s, 1 H), 8.28 (d, / = 2.3 Hz, 1 H), 8.24 (d, / = 8.5 Hz, 1 H), 8.18 (dd, / = 8.6, 2.3 Hz, 1 H), 8.07 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.5, 1.5 Hz, 1 H), 7.12 (d, / = 8.5 Hz, 1 H), 4.33-3.94 (m, 4 H), 3.90 (s, 3 H), 3.86-3.74 (m, 4 H), 3.35-3.10 (m, 3 H), 1.68 (s, 2 H), 1.59 (dq, / = 14.0, 7.2 Hz, 2 H), 1.14 (d, / = 6.0 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) nt/z 511.2 [M + H+] with a purity of >99%. Propyl ( ?)-4-(2-(4-(l-aminocyclopropyl)phenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine-l-carboxylate B281)
Figure imgf000300_0001
Compound B281 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine- 1 -carboxylate and 4-( 1 - ammocyclopropyl)pheny]boronic acid hydrochloride as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.35 (s, 1 H), 8.26 (d, / = 8.5 Hz, 1 H), 8.20 (d, / = 8.5 Hz, 2 H), 8.08 (d, / = 1.0 Hz, 1 H), 7.70 (dd, / = 8.5, 1.4 Hz, 1 H), 7.49 (d, / = 8.5 Hz, 2 H), 4.33-3.72 (m, 6 H), 3.37-3.10 (m, 3 H), 2.31 (s, 2 H), 1.66-1.52 (m, 2 H), 1.14 (d, / = 6.4 Hz, 3 H), 1.07-0.96 (m, 4 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 507.2 [M + H+] with a purity of >98%.
Propyl (/?)-4-(2-(4-(l-amino-2-methylpropan-2-yl)phenyl)-4-chloroquinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B282)
Figure imgf000300_0002
Compound B282 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and (4~(l -amino~2-rf:eihylpropari-2- yl)phenyl)boronic acid hydrochloride as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.34 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.21 (d, / = 8.5 Hz, 2 H), 8.09 (d, / = 1.1 Hz, 1 H), 7.70 (dd, / = 8.5, 1.6 Hz, 1 H), 7.54 (d, / = 8.5 Hz, 2 H), 4.33-3.72 (m, 6 H), 3.37-3.10 (m, 3 H), 2.75 (s, 2 H), 1.69-1.52 (m, 2 H), 1.30 (s, 6 H), 1.14 (d, / = 6.5 Hz, 5 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 523.3 [M + H+] with a purity of >96%.
Propyl (2/?)-4-(4-chloro-2-(4-(l-(pyrrolidin-l-yl)ethyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate (B283)
Figure imgf000300_0003
Compound B283 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate and 4-(! - pyrrolidifioetliy^phenylborofiic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.35 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.22 (d, / = 8.3 Hz, 2 H), 8.09 (d, / = 0.6 Hz, 1 H), 7.71 (dd, / = 8.6, 1.4 Hz, 1 H), 7.49 (d, / = 8.3 Hz, 2 H), 4.33-3.72 (m, 6 H), 3.36 (q, / = 6.3 Hz, 1 H), 3.32-3.10 (m, 3 H), 2.60-2.52 (m, 2 H), 2.42-2.33 (m, 2 H), 1.69 (s, 4 H), 1.64-1.54 (m, 2 H), 1.36 (d, / = 6.6 Hz, 3 H), 1.14 (d, / = 6.4 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 549.3 [M + H+] with a purity of >97%.
Propyl (2/?)-4-(4-chloro-2-(4-(l-(dimethylamino)ethyl)phenyl)quinoline-7-carbonyl)-2- methylpiperazine- 1-carboxylate B284)
Figure imgf000301_0001
Compound B284 was synthesized according to General Procedure L using propyl (R)-4-(2,4- dichloroquinoline-7-carbonyl)-2-methylpiperazine-l -carboxylate and { 4-[ I -
(dimethyl arnino)ethyl]pheny] J boronic acid hydrochloride as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.36 (s, 1 H), 8.27 (d, / = 8.5 Hz, 1 H), 8.23 (d, / = 8.2 Hz, 2 H), 8.09 (d, / = 0.9 Hz, 1 H), 7.71 (dd, / = 8.6, 1.2 Hz, 1 H), 7.48 (d, / = 8.3 Hz, 2 H), 4.33-3.72 (m, 6 H), 3.41 (q, / = 6.6 Hz, 1 H), 3.36-3.10 (m, 3 H), 2.16 (s, 6 H), 1.67-1.53 (m, 2 H), 1.33 (d, / = 6.7 Hz, 3 H), 1.14 (d, / = 6.8 Hz, 3 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 523.2 [M + H+] with a purity of >97%.
Propyl 4-(2-(4-(l-amino-2-methylpropan-2-yl)phenyl)-4-chloroquinoline-7-carbonyl)piperazine- 1-carboxylate (B285)
Figure imgf000301_0002
Compound B285 was synthesized according to General Procedure L using propyl 4-(2,4- dichloroquinoline-7-carbonyl)piperazine- 1 -carboxylate and (4-( 1 -amino-2-methylpropa« -2- yl)phenyl)boronic acid hydrochloride as starting materials.
LCMS (ESI-TOF) m/z 509.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(2-methyl-l-(methylamino)propan-2-yl)phenyl)quinoline-7- carbonyl)piperazine-l-carboxylate (B286)
Figure imgf000302_0001
Step 1 : Compound B285 (90 mg, 0.177 mmol) was dissolved in dichloromethane (2 mL) and triethylamine (50 μL·, 0.354 mmol, 2 equiv). The mixture was cooled to 0 °C before adding άι-tert- butyl dicarbonate (46.3 mg, 0.212 mmol, 1.2 equiv) was added. After stirring for 30 min at room temperature, the mixture was quenched by addition of saturated ammonium chloride. The organic layer was removed and the aqueous layer was extracted twice with dichloromethane. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (0-50% ethyl acetate/hexanes) to afford propyl 4-(2-(4-(l-((tert-butoxycarbonyl)amino)-2-methylpropan-2-yl)phenyl)-4- chloroquinoline-7-carbonyl)piperazine-l-carboxylate as a white solid (101.7 mg, 94%).
Step 2: The intermediate above (100 mg, 0.164 mmol) was dissolved in N,N-dimethylformamide (1.5 mL) and cooled to 0 °C. Upon addition of sodium hydride, 60% dispersion in mineral oil (7.8 mg, 0.197 mmol, 1.2 equiv), the mixture was allowed to stir at room temperature for 30 min. Iodomethane (20 nL, 0.32 mmol, 2 equiv) was added dropwise and the mixture was allowed to stir for 2 h before quenching with saturated sodium bicarbonate. The aqueous layer was extracted with ethyl acetate and washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was used without further purification.
Step 3: The residue was dissolved in dichloromethane (0.1 mL) and trifluoroacetic acid (0.1 mL). After 10 min, the crude material was purified using preparative HPLC (35% acetonitrile/water; 0.1% formic acid) to afford compound B286 ) as a white solid (32 mg, 37%).
*H NMR (400 MHz, DMSO-<¾ δ 8.44 (s, 1 H), 8.27 (d, / = 8.6 Hz, 1 H), 8.24 (d, / = 8.5 Hz, 2 H), 8.12 (d, / = 1.2 Hz, 1 H), 7.74 (dd, / = 8.5, 1.5 Hz, 1 H), 7.55 (d, / = 8.5 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.75-3.37 (m, 8 H), 2.66 (s, 2 H), 2.24 (s, 3 H), 1.59 (dd, / = 11.9, 4.9 Hz, 2 H), 1.32 (s, 6 H), 0.89 (t, / = 7.4 Hz, 3 H).
LCMS (ESI-TOF) m/z 523.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-(4-(l-(dimethylamino)-2-methylpropan-2-yl)phenyl)quinoline-7- carbonyl)piperazine-l-carboxylate (B287)
Figure imgf000302_0002
Compound B285 (90 mg, 0.177 mmol) was dissolved in methanol (0.9 mL) and paraformaldehyde (106 mg) was added. After 90 min, sodium borohydride (33.5 mg, 0.885 mmol, 5 equiv) was added and the mixture was quenched with 2M hydrochloric acid after 30 min to pH 1. The mixture was purified by preparative HPLC (35% acetonitrile/water; 0.1 % formic acid) to give a mixture of compound 285, 286 and 287 as a white solid (19.1 mg). The mixture was re-dissolved in dichloromethane (0.3 mL) and triethylamine (5 μΐ., 0.036 mmol). Di-teri-butyl dicarbonate (6 mg, 0.027 mmol) was then added. After 30 min, the mixture was quenched with saturated ammonium chloride, and was extracted twice with dichloromethane. The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (0-4% methanol/dichloromethane) to afford compound B287 as white solid upon lyophilisation (12.1 mg, 13%).
*H NMR (400 MHz, DMSO-<¾ δ 8.45 (s, 1 H), 8.29-8.20 (m, 3 H), 8.12 (s, 1 H), 7.73 (dd, / = 8.5, 1.5 Hz, 1 H), 7.57 (d, / = 8.4 Hz, 2 H), 3.98 (t, / = 6.6 Hz, 2 H), 3.78-3.34 (m, 8 H), 2.02 (s, 6 H), 1.59 (dd, / = 13.5, 6.4 Hz, 2 H), 1.33 (s, 6 H), 0.89 (t, / = 6.9 Hz, 3 H).
LCMS (ESI-TOF) m/z 537.2 [M + H+] with a purity of >99%.
( ?)-5-(2-((/?)-4-(4-Chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carbonyl)-2-methylpiperazin- l-yl)-2-oxoethyl)pyrrolidin-2-one (D001)
Figure imgf000303_0001
Step 1 : Tert-butyl (R)-4-(2,4-dichloroquinoline-7-carbonyl)-2-methylpiperazine-l-carboxylate was synthesized according to General Procedure CI using compound S9 (where R1 = H) and tert-butyl (R)-2-methylpiperazine-l -carboxylate as starting materials.
Step 2: (R)-(2,4-Dichloroquinolin-7-yl)(3-methylpiperazin-l -yl)methanone was synthesized by subjecting the intermediate above to 1 : 1 trifluoroacetic acid/dichloromethane mixture for 10 min followed by a basic work-up. The crude material was used directly without further purification.
Step 3: (R)-5-(2-((R)-4-(2,4-Dichloroquinoline-7-carbonyl)-2-methylpiperazin-l -yl)-2- oxoethyl)pyrrolidin-2-one was synthesized according to General Procedure CI using (R)-2-(5- oxopyrrolidin-2-yl)acetic acid as starting material.
Step 4: Compound D001 was synthesized according to General Procedure L by using 1 - methylpyrrole-3-boronic acid pinacol ester as starting material.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.15 (d, / = 8.5 Hz, 1 H), 7.99 (s, 1 H), 7.89 (d, / = 0.7 Hz, 1 H), 7.65 (s, 1 H), 7.56 (dd, / = 8.4, 1.2 Hz, 1 H), 7.08 (s, 1 H), 6.84-6.72 (m, 2 H), 5.68 (s, 1 H), 4.62-3.51 (m, 8 H), 3.26 (s, 1 H), 2.25-1.98 (m, 3 H), 1.72-1.57 (m, 1H), 1.25 (s, 2 H), 1.13 (s, 3 H), 0.92-0.83 (m, 1 H). LCMS (ESI-TOF) nt/z 494.1 [M + H+] with a purity of >95%.
(4-Chloro-2-phenylquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole-4-carbonyl)piperazin-l- yl)methanone (D002)
Figure imgf000304_0001
Step 1 : 4-Chloro-2-phenylquinoline-7-carboxylic acid was synthesized according to General Procedure I and K using acetophenone as starting material.
Step 2: (l-Cyclopropyl-lH-l ,2,3-triazol-4-yl)(piperazin-l -yl)methanone was synthesized according to General Procedure CI using l -cyclopropyl-lH-l ,2,3-triazole-4-carboxylic acid and teri-butyl piperazine-l-carboxylate as starting materials, followed by treatment with 1 : 1 trifluoroacetic acid/dichloromethane for 10 min.
Step 3: Compound D002 was synthesized according to General Procedure CI using intermediates from Step 1 and Step 2 as coupling partners.
!H NMR (400 MHz, DMSO-<¾ δ 8.61 (s, 1 H), 8.49 (s, 1 H), 8.34 (dd, / = 7.9, 1.6 Hz, 2 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.18 (d, / = 1.2 Hz, 1 H), 7.79 (dd, / = 8.5, 1.6 Hz, 1 H), 7.63-7.53 (m, 3 H), 4.29- 3.40 (m, 9 H), 1.32-1.07 (m, 4 H).
LCMS (ESI-TOF) nt/z 487.2 [M + H+] with a purity of >99%.
(4-Chloro-2-(l-methyl-lH-pyrrol-3-yl)quinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole-4- carbonyl)piperazin-l-yl)methanone (D003)
Figure imgf000304_0002
Step 1 : 4-Chloro-2-(l-methyl-lH-pyrrol-3-yl)quinoline-7-carboxylic acid was synthesized according to General Procedure I and K using 3 -acetyl- 1-methylpyrrole as starting material.
Step 2: Compound D003 was synthesized according to General Procedure CI using intermediate 1 and (l-cyclopropyl-lH-l ,2,3-triazol-4-yl)(piperazin-l-yl)methanone as starting materials. *H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.15 (d, / = 8.6 Hz, 1 H), 8.07 (s, 1 H), 7.94 (d, / = 1.1 Hz, 1 H), 7.71 (t, / = 1.7 Hz, 1 H), 7.61 (dd, / = 8.5, 1.5 Hz, 1 H), 6.88-6.77 (m, 2 H), 4.30-3.40 (m, 12 H), 1.31-1.06 (m, 4 H).
LCMS (ESI-TOF) nt/z 490.2 [M + H+] with a purity of >99%.
( ?)-(4-Chloro-2-(l-methyl-lH-pyrrol-3-yl)quinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole-4- carbonyl)-3-methylpiperazin-l-yl)methanone (D004)
Figure imgf000305_0001
Step 1 : (R)-(l-Cyclopropyl-lH-l ,2,3-triazol-4-yl)(2-methylpiperazin-l -yl)methanone was synthesized according to General Procedure CI using l -cyclopropyl-lH-l ,2,3-triazole-4-carboxylic acid and tert- butyl (R)-3-methylpiperazine-l -carboxylate as starting materials, followed by treatment with 1 : 1 trifluoroacetic acid/dichloromethane for 10 min.
Step 2: Compound D004 was synthesized according to General Procedure CI using 4-chloro-2-(l- methyl-lH-pyrrol-3-yl)quinoline-7-carboxylic acid and intermediate from Step 1 as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.47 (s, 1 H), 8.15 (d, / = 8.5 Hz, 1 H), 7.99 (s, 1 H), 7.91 (d, / = 1.2 Hz, 1 H), 7.65 (t, / = 1.8 Hz, 1 H), 7.57 (dd, / = 8.5, 1.5 Hz, 1 H), 6.80 (dt, / = 4.5, 2.7 Hz, 2 H), 4.90 (br s, 1 H), 4.50 (br s, 1 H), 4.21-3.81 (m, 3 H), 3.71 (s, 3 H), 3.35 (br s, 2 H), 3.26-3.11 (m, 1 H), 1.30-1.05 (m, 7 H).
LCMS (ESI-TOF) nt/z 504.2 [M + H+] with a purity of >99%.
( ?)-(4-Chloro-2-phenylquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole-4-carbonyl)-3- methylpiperazin- 1 -yl)methanone (D005)
Figure imgf000305_0002
Compound D005 was synthesized according to General Procedure CI using 4-chloro-2- phenylquinoline-7-carboxylic acid and (R)-( 1 -cyclopropyl- 1 H- 1 ,2,3 -triazol-4-yl)(2-methylpiperazin- l-yl)methanone as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾) δ 8.46 (s, 1 H), 8.38 (s, 1 H), 8.32-8.26 (m, 3 H), 8.15-8.12 (m, 1 H), 7.75 (dd, / = 8.6, 1.3 Hz, 1 H), 7.61-7.49 (m, 3 H), 4.91 (br s, 1 H), 4.51 (br s, 1 H), 4.21- 3.70 (m, 3 H), 3.38 (br s, 2 H), 3.20 (s, 1 H), 1.34-1.07 (m, 7 H). LCMS (ESI-TOF) nt/z 501.2 [M + H+] with a purity of >99%.
( ?)-(4-Chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3- triazole-4-carbonyl)-3-methylpiperazin-l-yl)methanone (D006)
Figure imgf000306_0001
Step 1 : (R)-(l-Cyclopropyl-lH-l ,2,3-triazol-4-yl)(4-(2,4-dichloroquinoline-7-carbonyl)-2- methylpiperazin-l-yl)methanone was synthesized according to General Procedure CI using compound S9 (where R1 = H) and (R)-(l-cyclopropyl-lH-l ,2,3-triazol-4-yl)(2-methylpiperazin-l - yl)methanone as starting materials.
Step 2: Compound D006 was synthesized according to General Procedure L using the above intermediate and 4-(2-hydroxypropa«-2~yl)phenyIbororiic acid as starting materials.
*H NMR (400 MHz, 80 °C, DMSO-<¾ δ 8.47 (s, 1 H), 8.36 (s, 1 H), 8.28 (d, / = 8.5 Hz, 1 H), 8.22 (d, / = 8.5 Hz, 2 H), 8.12 (d, / = 1.0 Hz, 1 H), 7.73 (dd, / = 8.6, 1.4 Hz, 1 H), 7.65 (d, / = 8.5 Hz, 2 H), 5.00-4.80 (m, 2 H), 4.50 (br s, 1 H), 4.30-3.68 (m, 3 H), 3.37 (br s, 2 H), 3.20 (t, / = 13.3 Hz, 1 H), 1.50 (s, 6 H), 1.32-1.06 (m, 7 H).
LCMS (ESI-TOF) nt/z 559.2 [M + H+] with a purity of >98%.
(4-Chloro-2-phenylquinolin-7-yl)(4-(l-methyl-lH-l,2,3-triazole-4-carbonyl)piperazin-l- yl)methanone (D007)
Figure imgf000306_0002
Step 1 : (l-Methyl-lH-l ,2,3-triazol-4-yl)(piperazin-l -yl)methanone was synthesized according to General Procedure CI using l -methyl-lH-l ,2,3-trizole-4-carboxylic acid and teri-butyl piperazine-1- carboxylate as starting materials, followed by treatment with 1 : 1 trifluoroacetic acid/dichloromethane.
Step 2: Compound D007 was synthesized according to General Procedure CI using intermediate from Step 1 and 4-chloro-2-phenylquinoline-7-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.53 (s, 1 H), 8.50 (s, 1 H), 8.34 (dd, / = 7.7, 1.5 Hz, 2 H), 8.30 (d, / = 8.4 Hz, 1 H), 8.19 (d, / = 0.5 Hz, 1 H), 7.79 (dd, / = 8.6, 1.4 Hz, 1 H), 7.64-7.53 (m, 3 H), 4.30- 3.42 (m, 11 H).
LCMS (ESI-TOF) nt/z 461.1 [M + H+] with a purity of >98%. (l-(Tert-butyl)-lH-l,2,3-triazol-4-yl)(4-(4-chloro-2-phenylquinoline-7-carbonyl)piperazin-l- yl)methanone (D008)
Figure imgf000307_0001
Step 1 : (l-(reri-butyl)-lH-l ,2,3-triazol-4-yl)(piperazin-l -yl)methanone was synthesized according to General Procedure CI using l-(teri-butyl)-lH-l ,2,3-trizole-4-carboxylic acid and teri-butyl piperazine-l-carboxylate as starting materials, followed by treatment with 1 : 1 trifluoroacetic acid/dichloromethane .
Step 2: Compound D008 was synthesized according to General Procedure CI using intermediate from Step 1 and 4-chloro-2-phenylquinoline-7-carboxylic acid as starting materials.
!H NMR (400 MHz, DMSO-<¾ δ 8.67 (s, 1 H), 8.49 (s, 1 H), 8.34 (dd, / = 7.7, 1.6 Hz, 2 H), 8.30 (d, / = 8.5 Hz, 1 H), 8.19 (s, 1 H), 7.79 (dd, / = 8.5, 1.3 Hz, 1 H), 7.62-7.50 (m, 3 H), 4.34-3.44 (m, 8 H), 1.63 (s, 9 H).
LCMS (ESI-TOF) nt/z 503.2 [M + H+] with a purity of >99%.
(4-Chloro-2-(4-(2-hydroxypropan-2-yl)phenyl)quinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole- 4-carbonyl)piperazin-l-yl)methanone (D009)
Figure imgf000307_0002
D009
Compound D009 was synthesized according to General Procedure CI using ( 1 -cyclopropyl-l H- 1 ,2,3 - triazol-4-yl)(piperazin-l-yl)methanone and commercially available 9-chloro-5,6,7,8- tetrahydroacridine-3-carboxylic acid as starting materials.
¾ NMR (400 MHz, DMSO-<¾) δ 8.60 (s, 1 H), 8.20 (d, / = 8.5 Hz, 1 H), 7.98 (d, / = 0.9 Hz, 1 H), 7.68 (dd, / = 8.7, 1.4 Hz, 1 H), 3.87 (dd, / = 210.0, 94.7 Hz, 9 H), 3.06 (s, 2 H), 2.99 (s, 2 H), 1.90 (s, 4 H), 1.18 (dd, / = 39.4, 4.8 Hz, 4 H).
LCMS (ESI-TOF) nt/z 465.2 [M + H+] with a purity of >98%. (9-Chloro-6-(pyridin-2-yl)-5,6,7,8-tetrahydroacridin-3-yl)(4-(l yclopropyl-lH-l,2,3-triazole-4- carbonyl)piperazin-l-yl)methanone (D010)
Figure imgf000308_0001
Compound D010 was prepared according to General Procedure A, B, and CI using 3-(pyridin-3- yl)cyclohexanone (General Procedure A) and (l-cyclopropyl-lH-l,2,3-triazol-4-yl)(piperazin-l- yl)methanone (General Procedure CI) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.60 (s, 1 H), 8.54 (d, / = 3.9 Hz, 1 H), 8.23 (d, / = 8.4 Hz, 1 H), 8.02 (d, / = 1.0 Hz, 1 H), 7.78 (td, / = 7.7, 1.8 Hz, 1 H), 7.70 (dd, / = 8.6, 1.4 Hz, 1 H), 7.43 (d, / = 8.1 Hz, 1 H), 7.32-7.23 (m, 1 H), 3.87 (dd, / = 220.9, 106.0 Hz, 10 H), 3.06 (dd, / = 32.5, 25.9 Hz, 4 H), 2.27 (s, 1 H), 2.10 (s, 1 H), 1.31-1.01 (m, 4 H).
LCMS (ESI-TOF) m/z 542.2 [M + H+] with a purity of >99%.
(4-Chloro-2-phenylquinolin-7-yl)(4-(l-cyclopropyl-lH-pyrazole-4-carbonyl)piperazin-l- yl)methanone (D011)
Figure imgf000308_0002
Step 1 : (l-Cyclopropyl-lH-pyrazol-4-yl)(piperazin-l-yl)methanone was synthesized according to General Procedure CI using l-cyclopropyl-lH-pyrazole-4-carboxylic acid and teri-butyl piperazine- 1-carboxylate as starting materials, followed by treatment with 1 : 1 trifluoroacetic acid/dichloromethane .
Step 2: Compound D011 was synthesized according to General Procedure CI using intermediate from Step 1 and 4-chloro-2-phenylquinoline-7-carboxylic acid as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.49 (s, 1 H), 8.34 (dd, / = 7.8, 1.4 Hz, 2 H), 8.30 (d, / = 8.6 Hz, 1 H), 8.17 (d, / = 1.1 Hz, 1 H), 8.14 (s, 1 H), 7.78 (dd, / = 8.6, 1.5 Hz, 1 H), 7.68 (s, 1 H), 7.62-7.51 (m, 3 H), 3.81-3.38 (m, 9 H), 1.14-0.87 (m, 4 H).
LCMS (ESI-TOF) m/z 486.2 [M + H+] with a purity of >99%.
(2-(4-(l-Amino-2-methylpropan-2-yl)phenyl)-4-chloroquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3- triazole-4-carbonyl)piperazin-l-yl)methanone (D012)
Figure imgf000309_0001
Step 1 : (1 -Cyclopropyl- 1 H- 1 ,2,3 -triazol-4-yl)(4-(2,4-dichloroquinoline-7-carbonyl)piperazin- 1 - yl)methanone was synthesized according to General Procedure CI using S9 (where R1 = H) and (1- cyclopropyl-lH-l ,2,3-triazol-4-yl)(piperazin-l-yl)methanone as starting materials.
Step 2: Compound D012 was synthesized according to General Procedure L using the above intermediate and (4-(l-aniino-2-methylpropan-2-yl)phe»yl)boiOmc acid hydrochloride as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.45 (s, 1 H), 8.31-8.22 (m, 3 H), 8.16 (d, / = 1.1 Hz, 1 H), 7.77 (dd, / = 8.6, 1.5 Hz, 1 H), 7.54 (d, / = 8.5 Hz, 2 H), 4.25-3.96 (m, 3 H), 3.85-3.42 (m, 6 H), 2.71 (s, 2 H), 1.29 (s, 6 H), 1.26-1.06 (m, 4 H).
LCMS (ESI-TOF) nt/z 558.2 [M + H+] with a purity of >99%.
(2-(4-(l-Aminocyclopropyl)phenyl)-4-chloroquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole-4- carbonyl)piperazin-l-yl)methanone (D013)
Figure imgf000309_0002
Compound D013 was synthesized according to General Procedure L using the above intermediate and 4-(l -aminocyclopropy1)phet3ylbororac acid hydrochloride as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.60 (s, 1 H), 8.46 (s, 1 H), 8.31-8.20 (m, 3 H), 8.15 (d, / = 1.1 Hz, 1 H), 7.75 (dd, / = 8.5, 1.6 Hz, 1 H), 7.48 (d, / = 8.6 Hz, 2 H), 4.25-3.95 (m, 3 H), 3.88-3.45 (m, 6 H), 1.27-1.10 (m, 4 H), 1.10-0.97 (m, 4 H).
LCMS (ESI-TOF) nt/z 542.2 [M + H+] with a purity of >96%.
(2-(3-(Aminomethyl)-4-methoxyphenyl)-4-chloroquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3- triazole-4-carbonyl)piperazin-l-yl)methanone (D014)
Figure imgf000310_0001
Compound D014 was synthesized according to General Procedure L and then General Procedure D using 3-cyano-4-methoxyphenyl boronic acid as starting materials (General Procedure L).
*H NMR (400 MHz, DMSO-<¾) δ 8.61 (s, 1 H), 8.44 (s, 1 H), 8.33 (d, / = 2.2 Hz, 1 H), 8.25 (d, / =
8.6 Hz, 1 H), 8.22 (dd, / = 8.7, 2.4 Hz, 1 H), 8.14 (d, / = 1.1 Hz, 1 H), 7.73 (dd, / = 8.5, 1.6 Hz, 1 H), 7.13 (d, / = 8.7 Hz, 1 H), 4.27-3.98 (m, 3 H), 3.89 (s, 3 H), 3.85-3.61 (m, 6 H), 3.51 (br s, 2 H), 1.30-
1.07 (m, 4 H).
LCMS (ESI-TOF) m/z 546.2 [M + H+] with a purity of >98%.
Allyl 4-(4-chloro-2,3-dimethylquinoline-7-carbon l)piperazine-l-carboxylate (COOl)
Figure imgf000310_0002
Compound COOl was prepared from General Procedure A, B and C2 using 2-butanone and 2-amino- terephthalic acid (General Procedure A) and allyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾) δ 8.19 (d, / = 8.8 Hz, 1 H), 7.96 (s, 1 H), 7.67 (d, / = 8.8 Hz, 1 H), 5.94-5.92 (m, 1 H), 5.31-5.18 (m, 2 H), 4.55 (d, / = 5.2 Hz, 2 H), 3.71-3.32 (m, 8 H), 2.71 (s, 3 H), 2.55 (s, 3 H).
LCMS (ESI-TOF) m/z 388.2 [M + H+] with a purity of >96%.
Allyl 4-(9-chloro-2,3-dihydro-lH-cyclopenta[b]quinoline-6-carbonyl)piperazine-l-carboxylate (C002)
Figure imgf000310_0003
Compound C002 was prepared using cyclopentanone and 2-amino-terephthalic acid as starting materials for cyclization using conditions similar to General Procedure A and B. The resulting product was reacted with allyl piperazine-l-carboxylate according to General Procedure C2.
*H NMR (400 MHz, DMSO-d6) δ 8.18 (d, / = 8.8 Hz, 1 H), 7.99 (d, / = 1.2 Hz, 1 H), 7.67 (dd, / = 8.4, 1.6 Hz, 1 H), 5.96-5.89 (m, 1 H), 5.31-5.18 (m, 2 H), 4.55 (d, / = 4.8 Hz, 2 H), 3.68-3.41 (m, 8 H), 3.19-3.13(m, 4 H), 2.23-2.16 (m, 2 H). LCMS (ESI-TOF) nt/z 400.2 [M + H+] with a purity of >96%.
Propyl 4-(4-chloro-2,3-dimethylquinoline-7-carbonyl)piperazine-l-carboxylate (C003)
Figure imgf000311_0001
Compound C003 was prepared from General Procedure A, B and C2 using 2-butanone and 2-amino- terephthalic acid (General Procedure A) and n-propyl piperazine-l-carboxylate (General Procedure C2) as starting materials.
*H NMR (400 MHz, DMSO-<¾ δ 8.18 (d, / = 8.8 Hz, 1 H), 7.95 (s, 1 H), 7.66 (d, / = 0.8 Hz, 1 H), 3.98 (d, / = 6.8 Hz, 2 H), 3.71-3.32 (m, 8 H), 2.70 (s, 3 H), 2.55 (s, 3 H), 1.61-1.55 (m, 2 H), 0.89 (t, / = 6.8 Hz, 3 H).
LCMS (ESI-TOF) nt/z 390.4 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-2-ethylquinoline-7-carbonyl)piperazine- 1-carboxylate (C004)
Figure imgf000311_0002
Step 1 : A mixture of 3-amino benzoic acid (5.0 g, 36.4 mmol) and methyl propionylacetate (30 mL) was heated to 90 °C and stirred for 24 h. After completion, the reaction mixture was washed with pentane to afford the resulting 3-(l-methoxy-l -oxopentan-3-ylideneamino)benzoic acid as a pale brown solid.
Step 2: A mixture of above imine (12.0 g, 48.1 mmol) and diphenyl ether (10 mL/g) was heated to 300 °C for 5 h in a sealed tube. The reaction mass was cooled to room temperature and diluted with hexanes. The resultant solid was collected by filtration and washed with hexanes to afford a mixture of desired methyl 2-ethyl-4-oxo-l ,4-dihydroquinoline-7-carboxylate and its regioisomer as a brown gum.
Step 3: The above mixture (2.0 g, 8.65 mmol) and phosphorus oxychloride (5 mL/g) was heated at 100 °C for 4 h. The reaction mass was concentrated under reduced pressure and excess of cold water was added to the residue, stirred until a free solid was formed. The resultant solid was collected by filtration, washed with hexane and dried. The crude product was purified by column chromatography (ethyl acetate/petroleum ether) to afford methyl 4-chloro-2-ethylquinoline-7carboxylate as an off- white solid.
Step 4: To a well stirred solution of the above intermediate (200 mg, 0.808 mmol) in a mixture of methanol, tetrahydrofuran and water (1 : 1 :0.5 mL) was added lithium hydroxide monohydrate (136 mg, 3.23 mmol) and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure. The residue was taken in cold water and acidified with 6 M hydrochloric acid, and the precipitated solid was collected by filtration to afford 4-chloro-2- ethylquinoline-7-carboxylic acid as brown solid.
Step 5: The above acid was reacted with n-propyl piperazine-l-carboxylate according to General Procedure C2 to afford C004.
*H NMR (400 MHz, DMSO-<¾) δ 8.22 (d, / = 8.4 Hz, 1 H), 8.01 (d, / = 1.2 Hz, 1 H), 7.80 (s, 1 H), 7.69 (dd, / = 1.6, 8.4 Hz, 1 H), 3.97 (t, / = 6.6 Hz, 2 H), 3.72-3.32 (m, 8 H), 2.99-2.93 (m, 2 H), 1.61-1.55 (m, 2 H), 1.32 (t, / = 7.6 Hz, 3 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) m/z 390.2 [M + H+] with a purity of >99%.
Propyl 4-(4-chloro-3-ethylquinoline-7-carbon l)piperazine- 1-carboxylate (C005)
Figure imgf000312_0001
Step 1 : To a suspension of 3-aminobenzoic acid (1 g, 7.3 mmol) in ethanol (15 mL) was added diethyl 2-ethyl-3-oxosuccinate (1.58 g, 7.3 mmol) and the mixture was stirred at reflux temperature. After 72 h, another portion of diethyl 2-ethyl-3-oxosuccinate (0.79 g, 3.65 mmol) was added and the reflux was continued for another 24 h. The reaction mass was concentrated under reduced pressure and the residue was stirred with a solution of 10% methanol in dichloromethane. The solids were separated by filtration and the filtrate was concentrated to afford 3-(l-Ethoxy-3-(ethoxycarbonyl)-l-oxopent-2-en- 2-ylamino)benzoic acid as a brown colored solid.
Step 2: To a pre -heated dodecylbenzene (10 mL) was added the above intermediate (1 g , 2.98 mmol) at 250 °C. The resulting mixture was stirred at same temperature for 6 h and then high vacuum was applied for 5 min. The reaction mass was cooled to room temperature and diluted with hexanes (50 mL) and stirred for 10 min. The filtrate was separated from the residue and the residue was purified by flash chromatography followed by preparative -HPLC to afford ethyl 3-ethyl-4-oxo-l,4- dihydroquinoline-7-carboxylate as a brown solid.
Step 3: To a stirred solution of the above intermediate (50 mg, 0.20 mmol) in tetrahydrofuran/water (2: 1, 10 mL) was added lithium hydroxide monohydrate (25.7 mg, 0.61 mmol) and stirred at room temperature for 20 h. The reaction mass was concentrated and the residue was dissolved in cold water (5 mL) and the resulting solution was acidified with 2 M hydrochloric acid to a pH 4. The resultant solid was collected by filtration and washed with cold water, hexanes and dried to afford ethyl-4-oxo- 1 ,4-dihydroquinoline-7-carboxylic acid as an off-white solid.
Step 4: A mixture of the above acid (40 mg, 0.18 mmol) and phosphorus oxychloride (1 mL) was stirred at 100 °C. After 4 h, the reaction mass was concentrated under reduced pressure, and cold water (10 mL) was added to the residue. The resultant solids were collected by filtration and washed with cold water, hexanes and dried to afford 4-chloro-3-ethylquinoline-7-carboxylic acid as an off white solid.
Step 5: The above acid was reacted with n-propyl piperazine-l-carboxylate according to General Procedure C2 to afford C005. *H NMR (400 MHz, DMSO-<¾ δ 8.95 (s, 1 H), 8.28 (d, / = 8.4 Hz, 1 H), 8.07 (d, / 7.76 (dd, / = 2.0, 8.8 Hz, 1 H), 3.97 (t, / = 7.2 Hz, 2 H), 3.7-3.32 (m, 8 H), 2.97 (q, /
1.62-1.54 (m, 2 H), 1.28 (t, / = 7.6 Hz, 3 H), 0.89 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 390.3 [M + H+] with a purity of >98%.
Propyl 4-(4-chloro-3-cyano-2-meth erazine-l-carboxylate (C006)
Figure imgf000313_0001
Compound C006 was prepared using 2-aminoterphthalic acid and 3-oxobutyronitrile as reagents for quinoline synthesis. The conditions were similar to that reported in General Procedure I. The resulting intermediate was reacted with n-propyl piperazine-l -carboxylate using General Procedure C2 conditions.
*H NMR (400 MHz, DMSO-<¾ δ 8.30 (d, / = 8.8 Hz, 1 H), 8.06 (d, / = 1.6 Hz, 1 H), 7.80 (dd, / = 1.6, 8.4 Hz, 1 H), 3.95 (t, / = 6.6 Hz, 2 H), 3.70-3.25 (m, 8 H), 2.83 (s, 3 H), 1.60-1.50 (m, 2 H), 0.89 (t, / = 7.2 Hz, 3 H).
LCMS (ESI-TOF) nt/z 401.2 [M + H+] with a purity of >99%.
Propyl 3-(4-chloroquinoline-7-carbo .3.1]nonane-9-carboxylate (C007)
Figure imgf000313_0002
Step 1 : According to General Procedure CI , commercially available 4-chloroquinoline-7-carboxylic acid was reacted with teri-butyl 7,9-diazabicyclo[3.3.1]nonane-9-carboxylate to give teri-butyl 3-(4- chloroquinoline-7-carbonyl)-3,9-diazabicyclo[3.3.1]nonane-9-carboxylate.
Step 2: To a solution of the intermediate from above (91.5 mg, 0.22 mmol) in dichloromethane (0.8 mL) was added trifluoroacetic acid (0.37 mL, 4.835 mmol, 22 equiv). The resulting mixture was stirred for 4 h before concentrating under reduced pressure. The residue was dissolved in ethyl acetate and then washed with saturated sodium bicarbonate. The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give (3,9- diazabicyclo [3.3.1 ] nonan-3 -yl) (4-chloroquinolin-7 -yl)methanone .
Step 3: To a solution of the above residue (48.2 mg, 0.153 mmol) in dichloromethane (1.5 mL) was added triethylamine (0.043 mL, 0.308 mmol, 2 equiv) and propyl chloroformate (0.030 mL, 0.267 mmol, 1.7 equiv). The mixture was stirred for 30 min before quenching by the addition of saturated sodium bicarbonate. The aqueous layer was extracted 3 times with ethyl acetate and the combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude material was purified by column chromatography (ethyl acetate/hexanes) to afford C007 as a white solid (29 mg, 47%) upon lyophilization. *H NMR (400 MHz, DMSO-<¾ δ 8.92 (d, / = 4.7 Hz, 1 H), 8.32 (d, / = 8.6 Hz, 1 H), 8.05 (s, 1 H), 7.86 (d, / = 4.7 Hz, 1 H), 7.75 (d, / = 8.6 Hz, 1 H), 4.62 (d, / = 13.3 Hz, 1 H), 4.25 (s, 1 H), 4.02 (br s, 2 H), 3.62-3.45 (m, 2 H), 3.14-3.11 (m, 1 H), 2.12-2.02 (m, 2 H), 1.86-1.50 (m, 7 H), 0.93-0.85 (m, 3 H).
LCMS (ESI-TOF) nt/z 402.1 [M + H+] with a purity of >97%.
Propyl 4-(10-chloro-l,2,3,4-tetrahydrobenzo[b][l,6]naphthyridine-7-carbonyl)piperazine-l- carboxylate (C008)
Figure imgf000314_0001
Compound C008 was prepared using 2-aminoterphthalic acid and l-benzylpiperidin-4-one as starting materials for quinoline synthesis using similar conditions to General Procedure A and B. The resulting intermediate was reacted with n-propyl piperazine-l -carboxylate according to General Procedure C2. The resulting intermediate (0.7 g, 1.38 mmol) was dissolved in 1 ,2-dichloroethane (10 mL) and chloroethyl chloroformate (130.3 mg, 0.91 mmol) was added at 0 °C. The reaction mixture was then stirred at 80°C for 2 h. Upon cooling, the reaction mixture was concentrated under reduced pressure. To the resulting residue was added methanol (40 mL) and the mixture was stirred at 60 °C for 1 h. The reaction mixture was concentrated under reduced pressure and drops of concentrated hydrochloric acid were added and then partitioned between ethyl acetate and saturated sodium carbonate solution. The combined organic layer was washed with water and brine, dried over anhydrous sodium sulfate, filtered, concentrated. The crude material was purified by preparative - HPLC to afford C008.
*H NMR (400 MHz, CDC13) δ 8.25 (d, / = 8.0 Hz, 1 H), 7.99 (s, 1 H), 7.61 (dd, / = 8.8, 1.6 Hz, 1 H), 4.29 (s, 2 H), 4.08 (t, / = 6.4 Hz, 2 H), 3.82-3.48 (m, 8 H), 3.33-3.30 (m, 2 H), 3.18-3.15 (m, 2 H), 1.69-1.64 (m, 3 H), 0.95 (t, / = 7.6 Hz, 3 H).
LCMS (ESI-TOF) nt/z 417.2 [M + H+] with a purity of >95%.
(2-(3-(Aminomethyl)-4-fluorophenyl)-4-chloroquinolin-7-yl)(4-(l-cyclopropyl-lH-l,2,3-triazole- 4-carbonyl)piperazin-l-yl)m
Figure imgf000314_0002
Compound E019 was synthesized according to General Procedure L by using 2-iluoro-5- (tetrametiiyl-l,3,2-dioxaborolan-2-yl)phenyl3nietIianaitiine hydrochloride as starting material.
*H NMR (400 MHz, DMSO-<¾ δ 8.61 (s, ~l H), 8.50 (s, 1 H), 8.47 (dd, / = 7.4, 2.2 Hz, 1 H), 8.29 (d, / = 8.5 Hz, 1 H), 8.27-8.22 (m, 1 H), 8.18 (d, / = 1.2 Hz, 1 H), 7.78 (dd, / = 8.6, 1.5 Hz, 1 H), 7.33 (dd, / = 9.6, 8.8 Hz, 1 H), 4.25-3.96 (m, 3 H), 3.86 (s, 2 H), 3.84-3.60 (m, 4 H), 3.51 (s, 2 H), 2.02 (s, 1 H), 1.27-1.09 (m, 4 H).
LCMS (ESI-TOF) m/z 534.2 [M + H+] with a purity of >99%. (4-Chloro-2-(3-((dimethylamino)methyl)-4-fluorophenyl)quinolin-7-yl)(4-(l-cyclopropyl-lH- l,2,3-triazole-4-carbonyl)piperazin-l-yl)methanone (E020)
Figure imgf000315_0001
Compound E020 was synthesized according to General Procedure L by using 3-((dimethylannno) mei'hyl)-4-fhiorophenylboronic add hydrochloride as starting material.
*H NMR (400 MHz, DMSO-<¾ δ 8.60 (s, 1 H), 8.49 (s, 1 H), 8.38 (dd, / = 7.2, 2.3 Hz, 1 H), 8.32- 8.25 (m, 2 H), 8.19 (s, 1 H), 7.78 (dd, / = 8.6, 1.5 Hz, 1 H), 7.37 (dd, / = 9.4, 9.0 Hz, 1 H), 4.28-3.95 (m, 3 H), 3.85-3.61 (m, 4 H), 3.61-3.42 (m, 4 H), 2.22 (s, 6 H), 1.18 (d, / = 36.4 Hz, 4 H).
LCMS (ESI-TOF) nt/z 562.2 [M + H+] with a purity of >98%.
Comparative Example 1
The following compounds in Table 5 are disclosed in the prior art and were synthesized and tested. Table 4. Table showing the list of prior art compounds and their biological activity
Figure imgf000315_0002
X4 0 CI 6.2 0
known compound ethyl 4-(9-chloro-2,3-dihydro-lH- cyclopenta[b]quinoline-6-carbonyl)piperazine- 1 - carboxylate
X5 ND
known from Mitchell et al.
There was no reported biochemical assay data in Peserico et.al. for comparative compound XI, and comparative compound XI was found to be inactive against SMYD3.
Comparative compound X2 and X3 were found to act against a different target ubiquitin specific protease 7, but were not active against SMYD3.
Comparative compound X4 was found to be moderately active against SMYD3 (6.2 μΜ), but suffered from poor metabolic stability due to high metabolic clearance in the human/mouse liver microsomes stability tests (half-life, t1/2 = 9 min/ 4 min respectively). In addition, comparative compound X4 was found to have poor target engagement compared to a more advanced compound B019.
Comparative compound X5 was found to be very active against SMYD3 (3 nM) using the specified assay reported in the publication. Although the reported molecule was active against SMYD3, no antiproliferative cellular activity was disclosed. Moreover, the structure of the inhibitor is not related to the compounds in this application. Industrial Applicability
The compounds as defined above may find a multiple number of applications in which their ability to inhibit protein lysine methyltransf erases such as SMYD3. The compounds may also be used in treating or preventing a condition or disorder in a mammal in which inhibition of a protein methyl transferase and/or co-factor thereof and/or via an unspecified mechanism prevents, inhibits or ameliorates apathology or a symptomology of the condition. The condition or disorder may be cancer, angiogenic disorder or pathological angiogenesis, fibrosis and inflammatory conditions. The compounds may be particularly useful in treating cancer such as breast, gastric, pancreatic, colorectal, lung cancer and hepatocellular carcinoma and other hypervascular tumors as well as angiogenic diseases.
It will be apparent that various other modifications and adaptations of the invention will be apparent to the person skilled in the art after reading the foregoing disclosure without departing from the spirit and scope of the invention and it is intended that all such modifications and adaptations come within the scope of the appended claims.

Claims

Claims
1. A compound having the following Formula (I);
Figure imgf000318_0001
(I)
wherein
Z1 and Z2 are independently selected from O, S or NH;
X is a halogen;
R1 and R2 are independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
and wherein R1 and R2 may optionally be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S;
and wherein R1 and R2 may optionally form an optionally substituted aryl or optionally substituted heteroaryl together with the ring atoms that they are bonded to;
R3, R4, R5, R6, R7 and R8 are independently absent, or selected from the group consisting of a bond, H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
wherein any two of R3, R4, R5, R6, R7 or R8 may be taken together to form an optionally substituted cycloalkyl, an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S;
Y is selected from R9, OR9 or NHR9, wherein R9 is an optionally substituted C3 to C10 alkyl, optionally substituted C3 to C10 alkenyl, optionally substituted C3 to C10 alkynyl, optionally substituted C3 to C7 cycloalkyl, optionally substituted C2 to C10 haloalkyl, a substituted 5- membered heteroaryl comprising two to three heteroatoms selected from N, O or S or a Q to C2 alkyl substituted with an optionally substituted 5-membered heterocycloalkyl comprising one to two heteroatoms selected from N, O or S; or a pharmaceutically acceptable form or prodrug thereof.
2. The compound according to claim 1 , wherein X is chloro.
3. The compound according to claim 1 or 2, wherein Z1 is O.
4. The compound according to any of claims 1 to 3, wherein Z2 is O.
5. The compound according to any of claims 1 to 4, having the following Formula (II):
Figure imgf000319_0001
(II).
6. The compound according to any claims 1 to 5, wherein the optionally substituted alkyl is an optionally substituted C\-C\2 alkyl, the optionally substituted alkyloxy is an optionally substituted Ci- Ci6 alkyloxy, the optionally substituted cycloalkyl is an optionally substituted C3-C9 cycloalkyl, the optionally substituted heterocycloalkyl is an optionally substituted heterocycloalkyl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the optionally substituted aryl is an optionally substituted C6-C18 aryl, the optionally substituted heteroaryl is an optionally substituted heteroaryl having a ring atom number of 3 to 8 and having 1 to 3 heteroatoms independently selected from the group consisting of N, O and S, the optionally substituted alkenyl is an optionally substituted C2-Q2 alkenyl or the optionally substituted alkynyl is an optionally substituted C2-C12 alkynyl.
7. The compound according to any one of claims 1 to 6, wherein R9 is selected from a C3 to C10 alkyl, a C3 to C10 alkenyl, C2 to C10 haloalkyl, or a in each case C3 to C9 alkyl or C3 to C7 cycloalkyl substituted oxazolyl, isoxazolyl, 1 ,2-azole, pyrazolyl, triazolyl, or methylpyrrolidinonyl.
8. The compound according to any one of claims 1 to 7, having the following formula (Ila):
Figure imgf000319_0002
(Ila)
wherein A1 is O or NH, and
R10 is a Q to C9 alkyl or a C3 to C7 cycloalkyl.
9. The compound according to any of claims 1 to 7, wherein R9 is selected from the group consisting of propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, -CH2CH(CH3)2, -CH2CH=CH, 2- fluoroethyl, 3-fluoropropyl, 5-cyclopropylisoxazol-3-yl, 5-isobutylisoxazol-3-yl, 5-methylisoxazol-
3- yl 5-methylpyrazol-3-yl, 1 -methyl- 1, 2,3 -triazol-4-yl, l-cycloproyl-l,2,3-triazol-4-yl, 1-tert-butyl- l,2,3-triazol-4-yl, l-cyclopropyl-l,2-pyrazol-4-yl and (R)-pyrrolidin-2-onyl-5-methyl.
10. The compound according to any of claims 1 to 9, wherein R1 and R2 are independently selected from the group consisting of a bond, H, halogen, cyano, optionally substituted alkyl, optionally substituted phenyl, optionally substituted pyrazolyl, optionally substituted thiazolyl, optionally substituted thiophenyl, optionally substituted benzo[d]imidazolyl, optionally substituted indolyl, optionally substituted isoindolyl, optionally substituted indazolyl, optionally substituted pyrrolyl, optionally substituted pyridinyl, optionally substituted benzyl, optionally substituted benzo[d]dioxolyl, optionally substituted benzotriazolyl, optionally substituted benzoxazolyl, optionally substituted benzofuranyl, optionally substituted pyrazolopyridinyl, optionally substituted pyrrolopyrimidinyl, optionally substituted pyrrolopyridinyl, optionally substituted naphthyridinyl, optionally substituted pyrimidinyl, optionally substituted benzothiazolyl, optionally substituted cyclopropyl, an amino group optionally substituted with an optionally substituted phenyl and an amino group optionally substituted with an optionally substituted pyridinyl.
11. The compound according to any of claims 1 to 10, having the following formula (lib):
Figure imgf000320_0001
(lib).
12. The compound according to claim 11, wherein R1 is H or halogen, and R2 is selected from the group consisting of H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3-methylphenyl, 4- methylphenyl, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, 2-(trifluoromethyl)phenyl, 3- (trifluoromethyl)phenyl, 4-(trifiuoromethyl)phenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4- fluoromethylphenyl, 2-hydroxymethylphenyl, 3-hydroxymethylphenyl, 4-hydroxymethylphenyl, 2- methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphenyl, 3-ethoxyethylphenyl, 4- ethoxyethylphenyl, 2-(azidomethyl)phenyl, 3-(azidomethyl)phenyl, 4-(azidomethyl)phenyl, 2,3- difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyl, 3,5- difluorophenyl, 3,5-difluoro-4-hydroxyphenyl, 3,5-difluoro-4-(aminocarbonyl)phenyl, 3,5-difluoro-4- aminomethylphenyl, 2-cyanophenyl, 3-cyanophenyl, 4-cyanophenyl, 2-(cyanomethyl)phenyl, 3- (cyanomethyl)phenyl, 4-(cyanomethyl)phenyl, 2-nitrophenyl, 3-nitrophenyl, 4-nitrophenyl, 2- aminophenyl, 3-aminophenyl, 4-aminophenyl, 2-(aminomethyl)phenyl, 3-(aminomethyl)phenyl, 4- (aminomethyl)phenyl, 2-(dimethylamino)phenyl, 3-(dimethylamino)phenyl, 4-
(dimethylamino)phenyl, 2-(aminocarbonyl)phenyl, 3-(aminocarbonyl)phenyl, 4-
(aminocarbonyl)phenyl, 2-(methylaminocarbonyl)phenyl, 3-(methylaminocarbonyl)phenyl, 4- (methylaminocarbonyl)phenyl, 2-(ethylaminocarbonyl)phenyl, 3-(ethylaminocarbonyl)phenyl, 4- (ethylaminocarbonyl)phenyl, 4-(l-ethoxyethyl)phenyl, 4-(2-hydroxy-2-propyl)phenyl, 2-pyridinyl, 3- pyridinyl, 4-pyridinyl, 2-methyl-3-pyridinyl, 4-methyl-3-pyridinyl, 5-methyl-3-pyridinyl, 6-methyl-3- pyridinyl, 6-methoxycarbonyl-3 -pyridinyl, 2-thiophenyl, 3-thiophenyl, 1-pyrrolidinyl, 2-pyrrolidinyl,
3- pyrrolidinyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, l-methyl-3-pyrrolyl, 3-(l,2,5-trimethyl)-pyrrolyl, 2- ethynylphenyl, 3-ethynylphenyl, 4-ethynylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-(l- hydroxyethyl)phenyl, 3-(l-hydroxyethyl)phenyl, 4-(l-hydroxyethyl)phenyl, 2-(2- hydroxyethyl)phenyl, 3 -(2-hydroxyethyl)phenyl, 4-(2-hydroxyethyl)phenyl, 4-fluoro-3 -methylphenyl,
4- fluoro-2-methylphenyl, 3-fluoro-2-methylphenyl, 3-fluoro-4-methylphenyl, 3-fluoro-5- methylphenyl, 2-fluoro-5 -methylphenyl, 4-fluoro-3 -methoxyphenyl, 4-fluoro-2-methoxyphenyl, 3- fluoro-2-methoxyphenyl, 3-fluoro-4-methoxyphenyl, 3 -fluoro-5 -methoxyphenyl, 2-fluoro-5- methoxyphenyl, 4-fluoro-3-hydroxyphenyl, 4-fluoro-2-hydroxyphenyl, 4-hydroxy-3-fluorophenyl, 4- hydroxy-2 -fluorophenyl, 4-fluoro-3-hydroxymethylphenyl, 4-fluoro-2-hydroxymethylphenyl, 3- fluoro-2-hydroxymethylphenyl, 3-fluoro-4-hydroxymethylphenyl, 3-fluoro-5-hydroxymethylphenyl, 2-fluoro-5-hydroxymethylphenyl, 3-fluoro-4-(2-hydroxy-2-propyl)phenyl, 3-(aminocarbonyl)-4- fluorophenyl, 2-(aminocarbonyl)-4-fluorophenyl, 2-(aminocarbonyl)-3-fluorophenyl, 4- (aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-2- fluorophenyl, 4-fluoro-3-(methylaminocarbonyl)phenyl, 3-fluoro-4-(methylaminocarbonyl)phenyl, 4- fluoro-2-(methylaminocarbonyl)phenyl, 3-fluoro-2-(methylaminocarbonyl)phenyl, 4- (cyclopropylaminocarbonyl)phenyl, 2-(cyclopropylaminocarbonyl)phenyl, 3- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-3-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-4- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-2-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-2- (cyclopropylaminocarbonyl)phenyl, (3-fluoro-4-(dimethylaminocarbonyl)phenyl, 3-fluoro-5- (dimethylaminocarbonyl)phenyl, 2-fluoro-5-(dimethylaminocarbonyl)phenyl, 4-fluoro-3- (dimethylaminocarbonyl)phenyl, 4-fluoro-2-(dimethylaminocarbonyl)phenyl, 3 -fluoro-2- (dimethylaminocarbonyl)phenyl, 3-methyl-4-(methylaminocarbonyl)phenyl, 3-amino-4-fluorophenyl, 2-amino-4-fluorophenyl, 3-aminomethyl-4-fluorophenyl, 2-aminomethyl-4-fluorophenyl, 3- hydroxymethyl-4-methylphenyl, 2-hydroxymethyl-4-methyl -phenyl, 2-hydroxymethyl-3 -methylphenyl, 4-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-2- methylphenyl, 2-morpholinophenyl, 3-mo holinophenyl, 4-mo holinophenyl, 2-(pyrrolidin-l- yl)phenyl, 3-(pyrrolidin- 1 -yl)phenyl, 4-(pyrrolidin- 1 -yl)phenyl, 4-( 1 -amino- 1 -cyclopropyl)phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2- methylthiazolyl, 4-methylthiazolyl, 4-(dimethylamido)phenyl, 2-(dimethylamido)phenyl, 3- (dimethylamido)phenyl, 2-benzylamin, 3-benzylamin, 4-benzylamin, 2-methylaminophenyl, 3- methylaminophenyl, 4-methylaminophenyl, 6-(l-methyl)indazolyl, 6-(2-methyl)indazolyl, 5-(l- methyl)indazolyl, 4-(l-methyl)indazolyl, 3-(l-methyl)indazolyl, 5-(2-methyl)indazolyl, 5-(3- methyl)indazolyl, 5-(l-methyl)pyrazolyl, 4-(l-methyl)-pyrazolyl, 3-(l-methyl) pyrazolyl, 4-(l- isopropyl)-pyrazolyl, 4-( 1 -difluoromethyl)-pyrazolyl, 4-(5-trifluoromethyl)-pyrazolyl, 4-( 1 -(2,2,2)- trifluoroethyl)pyrazolyl, 4-(l-cyclopentyl)pyrazolyl, 2-(l-methyl) pyrazolyl-phenyl, 3-(l-methyl) pyrazolyl-phenyl, 4-(imidazol-l-yl)phenyl, 1 -imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-(4- methylpiperazino)phenyl, 3 -(4-methylpiperazino)phenyl, 2-(4-methylpiperazino)phenyl, 3 -[ 1 ,2,4] - triazol-4-ylphenyl, 2-[l,2,4]-triazol-4-yl phenyl, 4-[l,2,4]-triazol-4-ylphenyl, 3-(aminomethyl)-4- methoxyphenyl, 3-(aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-4-methoxyphenyl, 2- (aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-6-methoxyphenyl, 4-(aminomethyl)-3- methoxyphenyl, 2-(aminomethyl)-3-methoxyphenyl, 4-(dimethylaminomethyl)phenyl, 3- (dimethylaminomethyl)phenyl, 2-(dimethylaminomethyl)phenyl, 4-fluoro-3 - (dimethylaminomethyl)phenyl, 4-fluoro-2-(dimethylaminomethyl)phenyl, 4-methoxy-3 -methylphenyl, 2-methoxy-4-methylphenyl, 3-methoxy-5-methylphenyl, 3-methoxy-4-methylphenyl, 2-methoxy-5- methylphenyl, 2-methoxy-6-methylphenyl, 2-methoxy-3 -methylphenyl, 4-methoxy-3- hydroxymethylphenyl, 3-methoxy-4-hydroxymethylphenyl, 2-methoxy-4-hydroxymethylphenyl, 3- methoxy-5-hydroxymethylphenyl, 2-methoxy-5-hydroxymethylphenyl, 2-methoxy-6- hydroxymethylphenyl, 2-methoxy-3-hydroxymethylphenyl, 4-hydroxy-3-hydroxymethylphenyl, 4- hydroxy-3 -methylphenyl, 3-ethoxy-4-hydroxyphenyl, 3-hydroxy-4-methylphenyl, 2-hydroxy-4- methylphenyl, 3-cyano-4-methylphenyl, 4-cyano-3 -methylphenyl, 2-cyano-4-methylphenyl, 3-cyano- 5-methylphenyl, 2-cyano-5-methylphenyl, 2-cyano-6-methylphenyl, 2-cyano-3-methylphenyl, 4- (aminosulfonyl)phenyl, 3-(aminosulfonyl)phenyl, 2-(aminosulfonyl)phenyl, 3-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 3-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-6-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-3-methoxyphenyl, 4-(N,N- dimethylaminomethyl)-3-methoxyphenyl, 3-(morpholinomethyl)phenyl, 2-(mo holinomethyl)phenyl, 4-(morpholinomethyl)phenyl, 3-cyano-4-methoxyphenyl, 2-cyano-4-methoxyphenyl, 3-cyano-5- methoxyphenyl, 2-cyano-5-methoxyphenyl, 2-cyano-6-methoxyphenyl, 2-cyano-3-methoxyphenyl, 4- cyano-3-methoxyphenyl, 4-aminomethyl-3 -methylphenyl, 2- aminomethyl-4-methylphenyl, 3- aminomethyl-5 -methylphenyl, 3-aminomethyl-4-methylphenyl, 2-aminomethyl-5-methylphenyl, 2- aminomethyl-6-methylphenyl, 2-aminomethyl-3-methylphenyl, (l-methyl)cyclopropyl, (2- methyl)cyclopropyl, 1-fluorocyclopropyl, 4-(2-methyl)pyridinyl, 3-(4-methyl)-pyridinyl, 2-(4- methyl)-pyridinyl, 2-(5-methyl)-pyridinyl, 2-(6-methyl)-pyridinyl, 2-(3-methyl)-pyridinyl, 2-(3- acetamido)-pyridinyl, 2-(4-acetamido)-pyridinyl, 2-(5-acetamido)-pyridinyl, 2-(6-acetamido)- pyridinyl, 3-(2-acetamido)-pyridinyl, 3-(4-acetamido)-pyridinyl, 3-(5-acetamido)-pyridinyl, 3-(6- acetamido)-pyridinyl, 4-(2-acetamido)-pyridinyl, 4-(3-acetamido)-pyridinyl, 4-(N- methylsulfamoyl)phenyl, 3-(N-methylsulfamoyl)phenyl, 2-(N-methylsulfamoyl)phenyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 3- (N-methylsulfamoyl)pyrrolyl, 3-(N,N-dimethylsulfamoyl)pyrrolyl, 4-(N-methylamido)phenyl, 3-(N- methylamido)phenyl, 2-(N-methylamido)phenyl, 4-(N-methylaminomethyl)phenyl, 3-(N- methylaminomethyl)phenyl, 2-(N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)-4- methoxyphenyl, 3-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-4- methoxyphenyl, 2-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-6- methoxyphenyl, 4-(N-methylaminomethyl)-3-methoxyphenyl, 2-(N-methylaminomethyl)-3- methoxyphenyl, 4-(acetylamino)phenyl, 3-(acetylamino)phenyl, 2-(acetylamino)phenyl, and ethynyl, 2-(5-N,N-dimethylaminomethyl)thiophenyl, 5-(2-methyl)indazolyl, 5-(3-methyl)indazolyl, 5-(7- methyl)indazolyl, 5-lH-indazolyl, 6-lH-indazolyl, 3-(l-methyl)pyrrolyl, 3-(2- methoxycarbonyl)pyrrolyl, 4-(2-methoxy)pyridinyl, 4-(lH-pyrrolo[2,3-b]pyridinyl), 5-(lH- pyrrolo[2,3-b]pyridinyl), 2-methyl-5-(lH-pyrrolo[2,3-b]pyridinyl), 4-(pyrazol-l-yl)phenyl, 4-(lH- pyrazol-5-yl)phenyl, 4-(lH-pyrazol-4yl)phenyl, 4-(lH-pyrazol-3-yl)phenyl, 4-carboxy-3- methylphenyl, 3-lH-pyrazolyl, 4-lH-pyrazolyl, 5-1H pyrazolyl, 4-lH-benzimidazolyl, 5-1H- benzimidazolyl, l-methyl-5-benzimidazolyl, 2-methyl-5-lH-benzimidazolyl, l-methyl-6- benzimidazolyl, 2-hydroxy-5-lH-benzimidazolyl, 5-(2-methyl)-benzoxazolyl, 5-(l-methyl)indolyl, 5- (3-methyl)indolyl, 4-lH-indazolyl, 3-(hydroxymethyl)phenyl, 3-hydroxyphenyl, l,3-benzodioxol-5-yl and l,2,3-benzotriazol-6-yl, 3-methyl-5-(lH-pyrazolo[3,4-b]pyridinyl, l-methyl-5-(lH-pyrrazolo[3,4- b]pyridinyl, 2-amino-5-pyrimidinyl, l,5-naphthyl-3-yl, l,5-naphthyridin-3-yl, 5-benzofuranyl, 6-(2- methyl) -benzothiazolyl, 5-(2-methyl)-benzothiazolyl, 5-benzoxazolyl, 6-benzoxazolyl, 6-(2-methyl)- benzoxazolyl, 5-(2-methyl)-benzoxazolyl, 4-((2-methoxyethoxy)methyl)phenyl, 4-
(cyclopropylmethoxy)methyl)phenyl, 3-(2-(aminomethyl)-l,5-dimethyl)-pyrrolyl, 5-oxoisoindolinyl,
3- fluoro-4-pyrrolidin- 1 -yl-phenyl, 4-( 1 -aminocarbonylmethyl)-pyrazolyl, 4-(l -oxetan-3-yl)-pyrazolyl,
4- ( 1 -amino-2-methyl-2-propyl)phenyl, 4- 1 -(pyrrolidin- 1 -yl)ethyl)phenyl, 4-( 1 - dimethylamino)ethyl)phenyl, 4-(2-hydroxypropan-2-yl)phenyl, 4-(2-methyl, 1-methylamino-propan- 2-yl)phenyl, 4-(2-methyl, l-dimethylamino-propan-2-yl)phenyl, 4-(l-amino-2-hydroxypropan-2- yl)phenyl and 3-dimethylaminoethyl-4-methoxyphenyl.
13. The compound according to any one claims 1 to 12, having the following Formula (III):
Figure imgf000323_0001
(III).
wherein R1 and Rl la are independently selected from the group consisting of H, halogen, cyano, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl;
Rnb is absent, H or optionally substituted alkyl;
A2 is selected from CH, N, O or S; and
p is an integer selected from from 0, 1 or 2.
14. The compound according to claim 13, having the following Formula (Ilia):
Figure imgf000323_0002
(Ilia).
15. The compound according to claim 13 or 14, wherein R1 and Rl la are independently selected from the group consisting of a bond, H, cyano, methyl, ethyl, ethynyl, phenyl, 2-methylphenyl, 3- methylphenyl, 4-methylphenyl, 2-hydroxyphenyl, 3-hydroxyphenyl, 4-hydroxyphenyl, 2- (trifluoromethyl)phenyl, 3-(trifluoromethyl)phenyl, 4-(trifluoromethyl)phenyl, 2-chlorophenyl, 3- chlorophenyl, 4-chlorophenyl, 2-fluorophenyl, 3-fluorophenyl, 4-fluorophenyl, 2-fluoromethylphenyl, 3-fluoromethylphenyl, 4-fluoromethylphenyl, 2-hydroxymethylphenyl, 3-hydroxymethylphenyl, 4- hydroxymethylphenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2-ethoxyethylphenyl, 3-ethoxyethylphenyl, 4-ethoxyethylphenyl, 2-(azidomethyl)phenyl, 3-(azidomethyl)phenyl, 4- (azidomethyl)phenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyl, 3,5-difluorophenyl, 3,5-difluoro-4-hydroxyphenyl, 3,5-difluoro-4- (aminocarbonyl)phenyl, 3,5-difluoro-4-aminomethylphenyl, 2-cyanophenyl, 3-cyanophenyl, 4- cyanophenyl, 2-(cyanomethyl)phenyl, 3-(cyanomethyl)phenyl, 4-(cyanomethyl)phenyl, 2-nitrophenyl, 3-nitrophenyl, 4-nitrophenyl, 2-aminophenyl, 3-aminophenyl, 4-aminophenyl, 2- (aminomethyl)phenyl, 3-(aminomethyl)phenyl, 4-(aminomethyl)phenyl, 2-(dimethylamino)phenyl, 3- (dimethylamino)phenyl, 4-(dimethylamino)phenyl, 2-(aminocarbonyl)phenyl, 3-
(aminocarbonyl)phenyl, 4-(aminocarbonyl)phenyl, 2-(methylaminocarbonyl)phenyl, 3- (methylaminocarbonyl)phenyl, 4-(methylaminocarbonyl)phenyl, 2-(ethylaminocarbonyl)phenyl, 3- (ethylaminocarbonyl)phenyl, 4-(ethylaminocarbonyl)phenyl, 4-(l-ethoxyethyl)phenyl, 4-(2-hydroxy-
2- propyl)phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-methyl-3-pyridinyl, 4-methyl-3-pyridinyl, 5- methyl-3-pyridinyl, 6-methyl-3-pyridinyl, 6-methoxycarbonyl-3-pyridinyl, thiophenyls such as 2- thiophenyl, 3-thiophenyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-pyrrolyl, 2-pyrrolyl, 3- pyrrolyl, l-methyl-3-pyrrolyl, 3-(l,2,5-trimethyl)-pyrrolyl, 2-ethynylphenyl, 3-ethynylphenyl, 4- ethynylphenyl, 2-ethylphenyl, 3-ethylphenyl, 4-ethylphenyl, 2-(l -hydroxy ethyl)phenyl, 3-(l- hydroxyethyl)phenyl, 4-(l-hydroxyethyl)phenyl, 2-(2-hydroxyethyl)phenyl, 3-(2- hydroxyethyl)phenyl, 4-(2-hydroxyethyl)phenyl, 4-fluoro-3-methylphenyl, 4-fluoro-2-methylphenyl,
3- fluoro-2-methylphenyl, 3-fluoro-4-methylphenyl, 3-fluoro-5-methylphenyl, 2-fluoro-5- methylphenyl, 4-fluoro-3-methoxyphenyl, 4-fluoro-2-methoxyphenyl, 3-fluoro-2-methoxyphenyl, 3- fluoro-4-methoxyphenyl, 3-fluoro-5-methoxyphenyl, 2-fluoro-5-methoxyphenyl, 4-fluoro-3- hydroxyphenyl, 4-fluoro-2-hydroxyphenyl, 4-hydroxy-3 -fluorophenyl, 4-hydroxy-2-fluorophenyl, 4- fluoro-3-hydroxymethylphenyl, 4-fluoro-2-hydroxymethylphenyl, 3-fluoro-2-hydroxymethylphenyl, 3-fluoro-4-hydroxymethylphenyl, 3-fluoro-5-hydroxymethylphenyl, 2-fluoro-5-hydroxymethylphenyl, 3-fluoro-4-(2-hydroxy-2-propyl)phenyl, 3-(aminocarbonyl)-4-fluorophenyl, 2-(aminocarbonyl)-4- fluorophenyl, 2-(aminocarbonyl)-3-fluorophenyl, 4-(aminocarbonyl)-3-fluorophenyl, 5- (aminocarbonyl)-3-fluorophenyl, 5-(aminocarbonyl)-2 -fluorophenyl, 4-fluoro-3- (methylaminocarbonyl)phenyl, 3 -fluoro-4-(methylaminocarbonyl)phenyl, 4-fluoro-2- (methylaminocarbonyl)phenyl, 3-fluoro-2-(methylaminocarbonyl)phenyl, 4- (cyclopropylaminocarbonyl)phenyl, 2-(cyclopropylaminocarbonyl)phenyl, 3- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-3-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-4- (cyclopropylaminocarbonyl)phenyl, 4-fluoro-2-(cyclopropylaminocarbonyl)phenyl, 3-fluoro-2- (cyclopropylaminocarbonyl)phenyl, (3-fluoro-4-(dimethylaminocarbonyl)phenyl, 3-fluoro-5- (dimethylaminocarbonyl)phenyl, 2-fluoro-5-(dimethylaminocarbonyl)phenyl, 4-fluoro-3- (dimethylaminocarbonyl)phenyl, 4-fluoro-2-(dimethylaminocarbonyl)phenyl, 3 -fluoro-2- (dimethylaminocarbonyl)phenyl, 3-methyl-4-(methylaminocarbonyl)phenyl, 3-amino-4-fluorophenyl, 2-amino-4-fluorophenyl, 3-aminomethyl-4-fluorophenyl, 2-aminomethyl-4-fluorophenyl, 3- hydroxymethyl-4-methylphenyl, 2-hydroxymethyl-4-methyl -phenyl, 2-hydroxymethyl-3 -methyl- phenyl, 4-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-3-methylphenyl, 5-hydroxymethyl-2- methylphenyl, 2-morpholinophenyl, 3-morpholinophenyl, 4-mo holinophenyl, 2-(pyrrolidin-l- yl)phenyl, 3-(pyrrolidin- 1 -yl)phenyl, 4-(pyrrolidin- 1 -yl)phenyl, 4-( 1 -amino- 1 -cyclopropyl)phenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2- methylthiazolyl, 4-methylthiazolyl, 4-(dimethylamido)phenyl, 2-(dimethylamido)phenyl, 3- (dimethylamido)phenyl, 2-benzylamin, 3-benzylamin, 4-benzylamin, 2-methylaminophenyl, 3- methylaminophenyl, 4-methylaminophenyl, 6-(l-methyl)indazolyl, 6-(2-methyl)indazolyl, 5-(l- methyl)indazolyl, 5-(2-methyl)indazolyl, 4-(l-methyl)indazolyl, 3-(l-methyl)indazolyl, 5-(3- methyl)indazolyl, 5-(l-methyl)pyrazolyl, 4-(l-methyl)-pyrazolyl, 3-(l-methyl) pyrazolyl, 4-(l- isopropyl)-pyrazolyl, 4-( 1 -difluoromethyl)-pyrazolyl, 4-(5-trifluoromethyl)-pyrazolyl, 4-( 1 -(2,2,2)- trifluoroethyl)pyrazolyl, 4-(l-cyclopentyl)pyrazolyl, 2-(l-methyl) pyrazolyl-phenyl, 3-(l-methyl) pyrazolyl-phenyl, 4-(imidazol-l-yl)phenyl, 1-imidazolyl, 2-imidazolyl, 3-imidazolyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfonamoyl)phenyl,
4- (4-methylpiperazino)phenyl, 3-(4-methylpiperazino)phenyl, 2-(4-methylpiperazino)phenyl, 3- [l,2,4]-triazol-4-ylphenyl, 2-[l,2,4]-triazol-4-yl phenyl, 4-[l,2,4]-triazol-4-ylphenyl, 3- (aminomethyl)-4-methoxyphenyl, 3-(aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-4- methoxyphenyl, 2-(aminomethyl)-5-methoxyphenyl, 2-(aminomethyl)-6-methoxyphenyl, 4- (aminomethyl)-3-methoxyphenyl, 2-(aminomethyl)-3-methoxyphenyl, 4- (dimethylaminomethyl)phenyl, 3-(dimethylaminomethyl)phenyl, 2-(dimethylaminomethyl)phenyl, 4- fluoro-3-(dimethylaminomethyl)phenyl, 4-fluoro-2-(dimethylaminomethyl)phenyl, 4-methoxy-3- methylphenyl, 2-methoxy-4-methylphenyl, 3-methoxy-5-methylphenyl, 3-methoxy-4-methylphenyl, 2-methoxy-5 -methylphenyl, 2-methoxy-6-methylphenyl, 2-methoxy-3-methylphenyl, 4-methoxy-3- hydroxymethylphenyl, 3-methoxy-4-hydroxymethylphenyl, 2-methoxy-4-hydroxymethylphenyl, 3- methoxy-5-hydroxymethylphenyl, 2-methoxy-5-hydroxymethylphenyl, 2-methoxy-6- hydroxymethylphenyl, 2-methoxy-3-hydroxymethylphenyl, 4-hydroxy-3-hydroxymethylphenyl, 4- hydroxy-3 -methylphenyl, 3-ethoxy-4-hydroxyphenyl, 3-hydroxy-4-methylphenyl, 2-hydroxy-4- methylphenyl, 3-cyano-4-methylphenyl, 4-cyano-3 -methylphenyl, 2-cyano-4-methylphenyl, 3-cyano-
5- methylphenyl, 2-cyano-5-methylphenyl, 2-cyano-6-methylphenyl, 2-cyano-3-methylphenyl, 4- (aminosulfonyl)phenyl, 3-(aminosulfonyl)phenyl, 2-(aminosulfonyl)phenyl, 3-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 3-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-4-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-5-methoxyphenyl, 2-(N,N- dimethylaminomethyl)-6-methoxyphenyl, 2-(N,N-dimethylaminomethyl)-3-methoxyphenyl, 4-(N,N- dimethylaminomethyl)-3-methoxyphenyl, 3-(mo holinomethyl)phenyl, 2-(morpholinomethyl)phenyl, 4-(morpholinomethyl)phenyl, 3-cyano-4-methoxyphenyl, 2-cyano-4-methoxyphenyl, 3-cyano-5- methoxyphenyl, 2-cyano-5-methoxyphenyl, 2-cyano-6-methoxyphenyl, 2-cyano-3-methoxyphenyl, 4- cyano-3-methoxyphenyl, 4-aminomethyl-3 -methylphenyl, 2- aminomethyl-4-methylphenyl, 3- aminomethyl-5 -methylphenyl, 3-aminomethyl-4-methylphenyl, 2-aminomethyl-5-methylphenyl, 2- aminomethyl-6-methylphenyl, 2-aminomethyl-3-methylphenyl, (l-methyl)cyclopropyl, (2- methyl)cyclopropyl, 1-fluorocyclopropyl, 4-(2-methyl)pyridinyl, 3-(4-methyl)-pyridinyl, 2-(4- methyl)-pyridinyl, 2-(5-methyl)-pyridinyl, 2-(6-methyl)-pyridinyl, 2-(3-methyl)-pyridinyl, 2-(3- acetamido)-pyridinyl, 2-(4-acetamido)-pyridinyl, 2-(5-acetamido)-pyridinyl, 2-(6-acetamido)- pyridinyl, 3-(2-acetamido)-pyridinyl, 3-(4-acetamido)-pyridinyl, 3-(5-acetamido)-pyridinyl, 3-(6- acetamido)-pyridinyl, 4-(2-acetamido)-pyridinyl, 4-(3-acetamido)-pyridinyl, 4-(N- methylsulfamoyl)phenyl, 3-(N-methylsulfamoyl)phenyl, 2-(N-methylsulfamoyl)phenyl, 4-(N,N- dimethylsulfamoyl)phenyl, 3-(N,N-dimethylsulfamoyl)phenyl, 2-(N,N-dimethylsulfamoyl)phenyl, 3- (N-methylsulfamoyl)pyrrolyl, 3-(N,N-dimethylsulfamoyl)pyrrolyl, 4-(N-methylamido)phenyl, 3-(N- methylamido)phenyl, 2-(N-methylamido)phenyl, 4-(N-methylaminomethyl)phenyl, 3-(N- methylaminomethyl)phenyl, 2-(N-methylaminomethyl)phenyl, 3-(N-methylaminomethyl)-4- methoxyphenyl, 3-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-4- methoxyphenyl, 2-(N-methylaminomethyl)-5-methoxyphenyl, 2-(N-methylaminomethyl)-6- methoxyphenyl, 4-(N-methylaminomethyl)-3-methoxyphenyl, 2-(N-methylaminomethyl)-3- methoxyphenyl, 4-(acetylamino)phenyl, 3-(acetylamino)phenyl, 2-(acetylamino)phenyl, and ethynyl, 2-(5-N,N-dimethylaminomethyl)thiophenyl, 5-(2-methyl)indazolyl, 5-(3-methyl)indazolyl, 5-(7- methyl)indazolyl, 5-lH-indazolyl, 6-lH-indazolyl, 3-(l-methyl)pyrrolyl, 3-(2- methoxycarbonyl)pyrrolyl, 4-(2-methoxy)pyridinyl, 4-(lH-pyrrolo[2,3-b]pyridinyl), 5-(lH- pyrrolo[2,3-b]pyridinyl), 2-methyl-5-(lH-pyrrolo[2,3-b]pyridinyl), 4-(pyrazol-l-yl)phenyl, 4-(lH- pyrazol-5-yl)phenyl, 4-(lH-pyrazol-4yl)phenyl, 4-(lH-pyrazol-3-yl)phenyl, 4-carboxy-3- methylphenyl, 3-lH-pyrazolyl, 4-lH-pyrazolyl, 5-1H pyrazolyl, 4-lH-benzimidazolyl, 5-1H- benzimidazolyl, l-methyl-5-benzimidazolyl, 2-methyl-5-lH-benzimidazolyl, l-methyl-6- benzimidazolyl, 2-hydroxy-5-lH-benzimidazolyl, 5-(2-methyl)-benzoxazolyl, 5-(l-methyl)indolyl, 5- (3-methyl)indolyl, 4-lH-indazolyl, 3-(hydroxymethyl)phenyl, 3-hydroxyphenyl, l,3-benzodioxol-5-yl and l,2,3-benzotriazol-6-yl, 3-methyl-5-(lH-pyrazolo[3,4-b]pyridinyl, l-methyl-5-(lH-pyrrazolo[3,4- b]pyridinyl, 2-amino-5-pyrimidinyl, l,5-naphthyl-3-yl, l,5-naphthyridin-3-yl, 5-benzofuran, 6-(2- methyl)-benzothiazolyl, 5-(2-methyl)-benzothiazolyl, 5-benzoxazolyl, 6-benzoxazolyl, 6-(2-methyl)- benzoxazol, 4-((2-methoxyethoxy)methyl)phenyl, 4-(cyclopropylmethoxy)methyl)phenyl, 3-(2- (aminomethyl)-l,5-dimethyl)-pyrrolyl, 5-oxoisoindolinyl, 3-fluoro-4-pyrrolidin-l-yl-phenyl, 4-(l- aminocarbonylmethyl)-pyrazolyl, 4-(l -oxetan-3-yl)-pyrazolyl, 4-(l -amino-2-methyl-2-propyl)phenyl, 4- 1 -(pyrrolidin- 1 -yl)ethyl)phenyl, 4-( 1 -dimethylamino)ethyl)phenyl, 4-(2-hydroxypropan-2- yl)phenyl, 4-(2-methyl, l-methylamino-propan-2-yl)phenyl, 4-(2-methyl, l-dimethylamino-propan-2- yl)phenyl, 4-(l-amino-2-hydroxypropan-2-yl)phenyl and 3-dimethylaminoethyl-4-methoxyphenyl.
16. The compound according to any one of claims 1 to 9, wherein R1 and R2 are taken together to form an optionally substituted 5-membered cycloalkyl, an optionally substituted 6-membered cycloalkyl, an optionally substituted 5-membered heterocycloalkyl or an optionally substituted 6- membered heterocycloalkyl.
17. The com ound according to claim 16, having the following Formula (IV):
Figure imgf000326_0001
(IV)
wherein
A3 and A4 are independently selected from CH or N;
A5 and A6 are independently selected from CH, N, O or S;
R12a, R13a, R14 and R15 are independently selected from the group consisting of H, halogen, optionally substituted alkyl, optionally substituted alkene, optionally substituted alkyne, optionally substituted alkoxy, optionally substituted amino, optionally substituted acyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl and optionally substituted heteroaryl.
R12b and R13b are independently absent, H or an optionally substituted alkyl; and
q and r are independently integers selected from 0, 1 or 2.
18. The compound according to claim 17, wherein A3 and A4 are both C.
19. The compound according to claim 18, having the following Formula (IV a):
Figure imgf000327_0001
(IVa)
wherein R9 is an optionally substituted C3 to do alkyl, optionally substituted C3 to do alkenyl, optionally substituted to do alkynyl or optionally substituted d to d cycloalkyl.
20. The compound according to any one of claims 17 to 19, wherein R12a, R13a, R14 and R15 are independently selected from H, methyl ,ethyl, propyl, butyl, halogen, cyano, COOMe, COOEt, phenyl, 2-pyridinyl, 3-pyridinyl, 4-pyridinyl, 2-(3-methyl)pyridinyl, 2-(4-methyl)pyridinyl, 2-(5- methyl)pyridinyl, 2-(6-methyl)pyridinyl, 3-(2-methyl)pyridinyl, 3-(4-methyl)pyridinyl, 3-(5- methyl)pyridinyl, 3-(6-methyl)pyridinyl, 4-(2-methyl)pyridinyl, 4-(3-fluoro)pyridinyl, 2-(3- fluoro)pyridinyl, 2-(4-fluoro)pyridinyl, 2-(5-fluoro)pyridinyl, 2-pyrazinyl, 2-(6-fluoro)pyridinyl, 3-(2- fluoro)pyridinyl, 3-(4-fluoro)pyridinyl, 3-(5-fluoro)pyridinyl, 3-(6-fluoro)pyridinyl, 4-(2- fluoro)pyridinyl, 4-(3-fluoro)pyridinyl, 2-(3-cyano)pyridinyl, 2-(4-cyano)pyridinyl, 2-(5- cyano)pyridinyl, 2-(6-cyano)pyridinyl, 3-(2-cyano)pyridinyl, 3-(4-cyano)pyridinyl, 3-(5- cyano)pyridinyl, 3-(6-cyano)pyridinyl, 4-(2-cyano)pyridinyl, 2-[3-(aminocarbonyl)]pyridinyl, 2-[4- (aminocarbonyl)]pyridinyl, 2-[5-(aminocarbonyl)]pyridinyl, 2-[6-(aminocarbonyl)]pyridinyl, 3-[2- (aminocarbonyl)]pyridinyl, 3-[4-(aminocarbonyl)]pyridinyl, 3-[5-(aminocarbonyl)]pyridinyl, 3-[6- (aminocarbonyl)]pyridinyl, 4-[2-(aminocarbonyl)]pyridinyl, 2-[3-(aminomethyl)]pyridinyl, 2-[4- (aminomethyl)]pyridinyl, 2-[5-(aminomethyl)]pyridinyl, 2-[6-(aminomethyl)]pyridinyl, 3-[2- (aminomethyl)]pyridinyl, 3-[4-(aminomethyl)]pyridinyl, 3-[5-(aminomethyl)]pyridinyl, 3-[6- (aminomethyl)]pyridinyl, 4-[2-(aminomethyl)]pyridinyl, 2-pyrimidinyl, 5-pyrimidinyl, 4-pyrimidinyl, 4-(3-methyl)pyrimidyl, 2-thiazolyl, 3-thiazolyl, pyrrolidine, 5-methyl-l,2,4-oxadiazol-3-yl, NH2, N(CH3)2, CH2CH=CH2, CH=CH2, CH2N(CH3)2, CH2NH2, CH2CH2NH2, C(0)NH2, NHC(NH)NH2, CH2NHC(NH)NH2, 2-(4-ethynyl)pyridinyl, 3-(4-ethynyl)pyridinyl, 2-(6-ethynyl)pyridinyl, 2-(5- ethynyl)pyridinyl, 3-(4-ethynyl)pyridinyl, 3-(2-ethynyl)pyridinyl, 3-(5-ethynyl)pyridinyl, 3-(6- ethynyl)pyridinyl, 2-(3-cyano)pyrimidinyl, 2-(5-cyano)pyrimidinyl, 2-(6-cyano)pyrimidinyl, 6-(2- cyano)pyrimidinyl, 2-(l-methyl)pyrazolyl, 4-(l-methyl)pyrazolyl, CH2-pyrrolidine, CH2CH2- pyrrolidine, ethynyl.
21. The compound according to any one of claims 1 to 20, wherein R3, R4, R5, R6, R7 and R8 is independently selected from the group consisting of a bond, H, methyl, (S)-methyl, (R)-methyl, ethyl, (S)-ethyl, (R)-ethyl, cyano, -CH2OH, (S)-CH2OH, (R)-CH2OH, COOCH3, CH2OC(0)CH3, (R)- CH2OC(0)CH3, (S)-CH2OC(0)CH3, CH2OC(0)CH2CH2OCH3, (R)-CH2OC(0)CH2CH2OCH3, (S)- CH2OC(0)CH2CH2OCH3, CH2CH2OH, (R)-CH2CH2OH, (S)-CH2CH2OH, CH2OC(0)CH2CH2CH3, (R)-CH2OC(0)CH2CH2CH3, (S)-CH2OC(0)CH2CH2CH3, CF3, (R)-CF3, (S)-CF3, (S)-COOCH3, (R)- COOCH3, CH2OCH3, (S)-CH2OCH3, (R)-CH2OCH3, CONHCH3, (S)-CONHCH3, (R)-CONHCH3 CH2COOCH3, (S)-CH2COOCH3, (R)-CH2COOCH3, CH2OC(0)CH(CH3)2, (S)- CH2OC(0)CH(CH3)2,CH2CONHCH3, (S)-CH2CONHCH3, (R)-CH2CONHCH3> (R)- CH2OC(0)CH(CH3)2, CONH2, (S)-CONH2, (R)-CONH2, CH2CON(CH3)2, (S)-CH2CON(CH3)2, (R)- CH2CON(CH3)2 and CH2C(0)NH(CH3).
22. The compound according to claim 21, wherein R4 and R5 or R6 and R7 may be taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S.
23. The compound according to claim 22, wherein R4 and R5 or R6 and R7 may be taken together to form a cyclopropane.
24. The compound according to any one of claims 1 to 23, wherein R3 and R4, R3 and R5, R3 and R6, R3 and R7, R3 and R8, R4 and R6, R4 and R7, R4 and R8, R5 and R6, R5 and R7, R5 and R8, R6 and R8 or R7 and R8 are taken together to form an optionally substituted alkylene bridge or an optionally substituted alkylene bridge wherein one or two alkylene units may be replaced with O, NH or S .
25. The compound according to claim 24, wherein the bridge of the optionally substituted alkylene bridge is a 1 -carbon, 2-carbon or 3-carbon alkylene bridging group wherein optionally one or two alkylene units are replaced with O, NH or S.
26. The compound according to any one of claims 1 to 25, selected from the group consisting of:
Figure imgf000328_0001
328
Figure imgf000329_0001
329
Figure imgf000330_0001
330
Figure imgf000331_0001
331
Figure imgf000332_0001
332
Figure imgf000333_0001
333
Figure imgf000334_0001
334
Figure imgf000335_0001
335
Figure imgf000336_0001
336
Figure imgf000337_0001
337
Figure imgf000338_0001
338
Figure imgf000339_0001
339
Figure imgf000340_0001
340
Figure imgf000341_0001
341
Figure imgf000342_0001
342
Figure imgf000343_0001
343
Figure imgf000344_0001
344
Figure imgf000345_0001
345
Figure imgf000346_0001
346
Figure imgf000347_0001
347
Figure imgf000348_0001
348
Figure imgf000349_0001
349
Figure imgf000350_0001
or a pharmaceutically acceptable form or prodrug thereof.
27. The compound according to any one of claims 1 to 26, wherein the compound is an enzyme inhibitor.
28. The compound according to claim 27, wherein the compound is a protein lysine methyltransferase (PKMT) inhibitor.
29. The compound according to claim 28, wherein the protein lysine methyltransferase is SMYD3.
30. The compound according to any one of claims 1 to 26, wherein the compound inhibits methylation of histone.
31. The compound according to claim 30, wherein the compound inhibits the trimethylation of histone H3 at lysine 4 (H3K4me3) and/or methylation of histone H4 at lysine 5 (H4K5me).
32. The compound according to any one of claims 1 to 26, wherein the compound modulates moystatin transcription and/or c-Met transcription.
33. The compound according to any one of claims 1 to 26, wherein the compound modulates the MEK-ERK mitogen-activated protein-kinase pathway.
34. The compound according to according to any one of claims 1 to 26, wherein the compound inhibits methylation of a lysine residue on MAP3K2.
35. The compound according to claim 34, wherein the lysine residue is K260.
36. A pharmaceutical composition comprising a compound of any one of claims 1 to 26, or a pharmaceutically acceptable form or prodrug thereof, and a pharmaceutically acceptable excipient.
37. A method of inhibiting SMYD3 in a cell comprising administering to a cell a compound of any one of claims 1 to 26, or a pharmaceutically acceptable form or prodrug thereof, or a composition according to claim 36.
38. The method according to claim 37, wherein the inhibition of SMYD3 further comprises the inhibition of cell proliferation.
39. The method according to claim 37 or 38, wherein the cell is in vitro.
40. The method according to claim 39, wherein the cell is from a cell line.
41. The method according to claim 40, wherein the cell line is an immortalized cell line, a genetically modified cell line or a primary cell line.
42. The method according to claim 40, wherein the cell line is selected from the group consisting of, HepG2, HCT116, A549, HPAF-II, CFPAC-1, HEK293, Hep3B, HuH7,SNU398, PLC/PRF/5, HuHl, Bel7404, HCCLM3, HLE, SK-HEP-1, Mahlavu, JHHl, JHH2, JHH4, JHH5, JHH7, SNU354, SNU368, SNU387, SNU423, SNU449, SNU739, SNU761, SNU886 and MIA PaCa-2.
43. The method according to claim 37 or 38 wherein the cell is from tissue of a subject.
44. The method according to claim 37 or 38, wherein the cell is in a subject.
45. A method of treating a SMYD-3-related disorder comprising administering to a subject in need of treatment a compound of any one of claims 1 to 26, or a pharmaceutically acceptable form or prodrug thereof, or a composition according to claim 36.
46. The method according to claim 45, wherein the disorder is cancer, angiogenic disorder or pathological angiogenesis, fibrosis or inflammatory conditions.
47. The method according to claim 46, wherein the disorder is lymphoma, cutaneous T-cell lymphoma, follicular lymphoma, or Hodgkin lymphoma, cervical cancer, ovarian cancer, breast cancer, lung cancer, prostate cancer, colorectal cancer, gastric cancer, pancreatic cancer, sarcoma, hepatocellular carcinoma, leukemia or myeloma, retinal angiogenic disease, liver fibrosis, kidney fibrosis, or myelofibrosis.
48. The method according to any one of claims 45 to 47, further comprising the step of administering an additional therapeutic agent in the subject.
49. The compound according to any one of claims 1 to 26, or a pharmaceutically form or prodrug thereof, or a composition according to claim 36 for use in therapy.
50. A use of a compound according to any one of claims 1 to 26, or a pharmaceutically acceptable form or prodrug thereof, or a composition according to claim 36, in the manufacture of a medicament for treatment of a SMYD3-related disorder.
51. The use according to claim 50, wherein the disorder is cancer, angiogenic disorder or pathological angiogenesis, fibrosis or inflammatory conditions.
52. The use according to claim 51, wherein the disorder is lymphoma, cutaneous T-cell lymphoma, follicular lymphoma, or Hodgkin lymphoma, cervical cancer, ovarian cancer, breast cancer, lung cancer, prostate cancer, colorectal cancer, gastric cancer, pancreatic cancer, sarcoma, hepatocellular carcinoma, leukemia or myeloma, retinal angiogenic disease, liver fibrosis, kidney fibrosis, or myelofibrosis.
53. The use according to any one of claims 50 to 52, wherein the medicament is administered with an additional therapeutic agent, wherein said medicament is to be administered in combination or alteration with the additional therapeutic agent.
54. A compound according to any one of claims 1 to 26, or a pharmaceutically acceptable form or prodrug thereof, or a composition according to claim 36, for use in the treatment of a SMYD3-related disorder.
55. The compound according to claim 54, wherein the disorder is cancer, angiogenic disorder or pathological angiogenesis, fibrosis or inflammatory conditions.
56. The compound according to claim 55, wherein the disorder is lymphoma, cutaneous T- cell lymphoma, follicular lymphoma, or Hodgkin lymphoma, cervical cancer, ovarian cancer, breast cancer, lung cancer, prostate cancer, colorectal cancer, gastric cancer, pancreatic cancer, sarcoma, hepatocellular carcinoma, leukemia or myeloma, retinal angiogenic disease, liver fibrosis, kidney fibrosis, or myelofibrosis
57. The compound according to any one of claims 54 to 56, wherein the compound is administered with an additional therapeutic agent, wherein said medicament is to be administered in combination or alteration with the additional therapeutic agent.
58. A process for synthesizing the compound according to claim 11 having the following Formula (III), comprising the steps of:
Figure imgf000353_0001
(III)
(a) contacting an optionally substituted aminobenzoate ester with a compound having the following Formula (Va) to form a cyclized product;
Figure imgf000353_0002
wherein R16 is selected from the group consisting of H, methyl, COOMe and COOEt;
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) selectively hydrolyzing the ester of the cyclized product of step (a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form the compound of formula (III);
Figure imgf000353_0003
wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
59. A process for synthesizing the compound according to claim 17 having the following formula (III), wherein R1 is optionally a halogen or hydrogen, comprising the steps of;
Figure imgf000354_0001
(HI),
(a) contacting an optionally substituted aminobenzoate ester with a compound having the following Formula (Vb) and phosphorus oxychloride to form a halogenated cyclized product;
Figure imgf000354_0002
(Vb);
(b) selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid and selectively functionalizing the carboxylic acid with a group having the following formula (VI) under reaction conditions to form an amide;
Figure imgf000354_0003
(VI);
(c) selectively functionalizing at least one halogen of the halogenated cyclized product of step (a) with a group having the following formula (Vila) or formula (Vllb) under reaction conditions to form the com ound of formula (III);
Figure imgf000354_0004
(Vila) (Vllb)
wherein step (b) and (c) may be performed simultaneously, sequentially or in any order.
60. A process for synthesizing the compound according to claim 17 having the following formula (IV), comprising the steps of;
Figure imgf000355_0001
(IV)
(a) contacting an amino substituted terephthalic acid or an ester thereof; with an optionally substituted cyclic ketone having the following Formula (VIII) to form a cyclized product;
Figure imgf000355_0002
(VIII)
(b) selectively displacing at least one ketone of the cyclized product of step (a) with a halogen;
(c) optionally selectively hydrolyzing the ester of the cyclized product of step a) to a carboxylic acid;
(d) selectively functionalizing the carboxylic acid of the cyclized product of step (a) or (c) with a group having the following formula (VI) under reaction conditions to form the compound of formula (V);
Figure imgf000355_0003
(VI)
wherein step (b), (c) and (d) may be performed simultaneously, sequentially or in any order.
61. The process according to claim 60, comprising the step of optionally hydrolyzing the carboxylic acid ester after formation of the cyclized product in step (a).
PCT/SG2016/050499 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics WO2017061957A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CA3001452A CA3001452A1 (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics
CN201680071788.2A CN108419435A (en) 2015-10-09 2016-10-10 Compound and epigenetics for treating cancer
SG11201802952TA SG11201802952TA (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics
JP2018518408A JP2018534276A (en) 2015-10-09 2016-10-10 Compounds for cancer treatment and epigenetics
AU2016335043A AU2016335043A1 (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics
KR1020187012151A KR20180061316A (en) 2015-10-09 2016-10-10 Compounds for the treatment and prophylactic genetics of cancer
BR112018007128A BR112018007128A2 (en) 2015-10-09 2016-10-10 compounds for cancer inhibition and epigenesis
EP16854005.2A EP3371151A4 (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics
US15/767,129 US20190071416A1 (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SG10201508413Q 2015-10-09
SG10201508413Q 2015-10-09

Publications (1)

Publication Number Publication Date
WO2017061957A1 true WO2017061957A1 (en) 2017-04-13

Family

ID=58488141

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SG2016/050499 WO2017061957A1 (en) 2015-10-09 2016-10-10 Compounds for treatment of cancer and epigenetics

Country Status (10)

Country Link
US (1) US20190071416A1 (en)
EP (1) EP3371151A4 (en)
JP (1) JP2018534276A (en)
KR (1) KR20180061316A (en)
CN (1) CN108419435A (en)
AU (1) AU2016335043A1 (en)
BR (1) BR112018007128A2 (en)
CA (1) CA3001452A1 (en)
SG (1) SG11201802952TA (en)
WO (1) WO2017061957A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018151678A1 (en) * 2017-02-15 2018-08-23 Agency For Science, Technology And Research Compounds for treatment of cancer and epigenetics
WO2018195450A1 (en) * 2017-04-21 2018-10-25 Epizyme, Inc. Combination therapies with ehmt2 inhibitors
WO2020011607A1 (en) 2018-07-09 2020-01-16 Fondation Asile Des Aveugles Inhibition of prc2 subunits to treat eye disorders
WO2023014861A1 (en) * 2021-08-05 2023-02-09 Flare Therapeutics Inc. Pparg inverse agonists and uses thereof
US20230098872A1 (en) * 2021-03-26 2023-03-30 Cedilla Therapeutics, Inc. Tead inhibitors and uses thereof
US11820747B2 (en) 2021-11-02 2023-11-21 Flare Therapeutics Inc. PPARG inverse agonists and uses thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108623591B (en) * 2017-03-24 2021-08-17 中国海洋大学 Application of paninula compound
CN109956909B (en) * 2019-04-24 2022-04-12 上海卡洛化学有限公司 Preparation method of 2-cyanopiperazine-1-tert-butyl carboxylate
KR20230132725A (en) * 2022-03-08 2023-09-18 한국과학기술원 Composition for preventing or treating cancer comprising SETMAR inhibitor

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006098342A1 (en) * 2005-03-16 2006-09-21 Astellas Pharma Inc. Piperazinyl compounds
CA2613322A1 (en) 2005-07-01 2007-01-11 Yusuke Nakamura Methods of modulating smyd3 for treatment of cancer
WO2011086178A1 (en) 2010-01-15 2011-07-21 Hybrigenics Sa Amidoacridine derivatives useful as selective inhibitors of ubiquitin specific protease 7
US8354223B2 (en) 2007-06-14 2013-01-15 Oncotherapy Science, Inc. Methods of identifying agents that modulate methylation of VEGFR1 by SMYD3

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5432696A (en) * 1995-04-07 1996-10-23 Pharmacopeia, Inc. Carbonyl-piperazinyl and piperidinil compounds which inhibit farnesyl protein transferase
ES2739621T3 (en) * 2013-10-15 2020-02-03 Janssen Pharmaceutica Nv Secondary alcohol modulators of ROR Gamma T of quinolinyl type
JP6441947B2 (en) * 2014-01-31 2018-12-19 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Kinoline-based kinase inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006098342A1 (en) * 2005-03-16 2006-09-21 Astellas Pharma Inc. Piperazinyl compounds
CA2613322A1 (en) 2005-07-01 2007-01-11 Yusuke Nakamura Methods of modulating smyd3 for treatment of cancer
US8354223B2 (en) 2007-06-14 2013-01-15 Oncotherapy Science, Inc. Methods of identifying agents that modulate methylation of VEGFR1 by SMYD3
WO2011086178A1 (en) 2010-01-15 2011-07-21 Hybrigenics Sa Amidoacridine derivatives useful as selective inhibitors of ubiquitin specific protease 7

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Prodrugs: Challenges and Rewards (Parts 1 and 2", 2007, SPRINGER
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING CO.
ARROWSMITH, C. H. ET AL.: "Epigenetic protein families: a new frontier for drug discovery", NAT. REV. DRUG DISCOV., vol. 11, 2012, pages 384 - 400, XP055329076, doi:10.1038/nrd3674
COPELAND, R. A.: "Molecular pathways: protein methyltransferases in cancer", CLIN. CANCER RES., vol. 19, 2013, pages 6344 - 6350
F.J. LEINWEBER, DRUG METAB. RES., vol. 18, 1987, pages 379
HAMAMOTO, R. ET AL.: "SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells", NAT. CELL BIOL., vol. 6, 2004, pages 731 - 740, XP002385375, doi:10.1038/ncb1151
MAZUR, P.: "SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer", NATURE, vol. 510, 2014, pages 283 - 287
MITCHELL, L. H. ET AL.: "Novel oxindole sulfonamides and sulfamides: EPZ031686, the first orally bioavailable small molecule SMYD3 inhibitor''.", ACS MED. CHEM. LETT., vol. 7, no. 2, 27 August 2015 (2015-08-27), pages 134 - 138, XP055376213, [retrieved on 20161123] *
MITCHELL, L. H.: "Novel Oxindole Sulfonamides and Sulfamides: EPZ031686, the First Orally Bioavailable Small Molecule SMYD3 Inhibitor", ACSMED.CHEM. LETT., 2015
PESERICO, A. ET AL.: "A SMYD3 small-molecule inhibitor impairing cancer cell growth''.", J. CELL. PHYSIOL., vol. 230, no. 10, 23 June 2015 (2015-06-23), pages 2447 - 2460, XP055376212, [retrieved on 20161123] *
PESERICO, A.: "A SMYD3 small-molecule inhibitor impairing cancer cell growth", J. CELL PHYSIOL., vol. 230, 2015, pages 2447 - 2460, XP055376212, doi:10.1002/jcp.24975
See also references of EP3371151A4 *
VAN ELLER ET AL.: "Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation", EPIGENETICS, vol. 7, 2012, pages 340 - 343

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018151678A1 (en) * 2017-02-15 2018-08-23 Agency For Science, Technology And Research Compounds for treatment of cancer and epigenetics
WO2018195450A1 (en) * 2017-04-21 2018-10-25 Epizyme, Inc. Combination therapies with ehmt2 inhibitors
CN110621316A (en) * 2017-04-21 2019-12-27 Epizyme股份有限公司 Combination therapy with EHMT2 inhibitors
US11672800B2 (en) 2017-04-21 2023-06-13 Epizyme, Inc. Combination therapies with EHMT2 inhibitors
CN110621316B (en) * 2017-04-21 2024-01-26 Epizyme股份有限公司 Combination therapy with EHMT2 inhibitors
WO2020011607A1 (en) 2018-07-09 2020-01-16 Fondation Asile Des Aveugles Inhibition of prc2 subunits to treat eye disorders
US20230098872A1 (en) * 2021-03-26 2023-03-30 Cedilla Therapeutics, Inc. Tead inhibitors and uses thereof
WO2023014861A1 (en) * 2021-08-05 2023-02-09 Flare Therapeutics Inc. Pparg inverse agonists and uses thereof
US11820747B2 (en) 2021-11-02 2023-11-21 Flare Therapeutics Inc. PPARG inverse agonists and uses thereof

Also Published As

Publication number Publication date
BR112018007128A2 (en) 2018-11-06
CN108419435A (en) 2018-08-17
EP3371151A1 (en) 2018-09-12
AU2016335043A1 (en) 2018-04-26
SG11201802952TA (en) 2018-05-30
EP3371151A4 (en) 2019-03-27
JP2018534276A (en) 2018-11-22
CA3001452A1 (en) 2017-04-13
KR20180061316A (en) 2018-06-07
US20190071416A1 (en) 2019-03-07

Similar Documents

Publication Publication Date Title
WO2017061957A1 (en) Compounds for treatment of cancer and epigenetics
JP6949952B2 (en) Spiro ring compound
JP6802251B2 (en) Cyanopyrrolidine as a DUB inhibitor for the treatment of cancer
RU2394825C2 (en) Pyrrolopyrazines suitable as aurora a kinase inhibitors
EP3197883B1 (en) Novel compounds
RU2717238C2 (en) Compounds of 1-cyano-pyrrolidines as usp30 inhibitors
TW201938561A (en) Chemical compounds
JP7373992B2 (en) Substituted pyrazole compounds and methods of their use for the treatment of hyperproliferative diseases
AU2008225766B2 (en) Organic compounds and their uses
AU2008258366B2 (en) 2-oxo-3-benzyl-benzoxazol-2-one derivatives and related compounds as Met kinase inhibitors for the treatment of tumors
CN109963842A (en) Benzimidazoles compound kinase inhibitor and its preparation method and application
WO2017007689A1 (en) Cot modulators and methods of use thereof
AU2017226004A1 (en) Inhibitors of WDR5 protein-protein binding
WO2022132914A1 (en) Prmts inhibitors
JP2008520738A5 (en)
AU2014354711A1 (en) Aminopyridine derivatives as TAM family kinase inhibitors
WO2015077193A1 (en) Inhibitors of lysine methyl transferase
JP2014513071A (en) Wind pathway blocker
KR20170036095A (en) 6-Alkynyl-pyridine derivatives as SMAC mimetics
KR20190098760A (en) Poly-ADP Ribose Polymerase (PARP) Inhibitor
EP2662367B1 (en) Novel indole or indazole derivative or salt thereof
TW202330537A (en) Wee1 inhibitor and the preparations and use thereof
BR112020026337A2 (en) TRICYCLIC COMPOUNDS
CA3227194A1 (en) Compounds for the treatment of pain, in particular neuropathic pain, and/or other diseases or disorders that are associated with at2r and/or at2r mediated signaling
BR112017005887B1 (en) COMPOUND, METHOD FOR PRODUCTION AND USE OF THE SAME, AS WELL AS PHARMACEUTICAL COMPOSITION

Legal Events

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

Ref document number: 16854005

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3001452

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 11201802952T

Country of ref document: SG

Ref document number: 2018518408

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018007128

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2016335043

Country of ref document: AU

Date of ref document: 20161010

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20187012151

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2016854005

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112018007128

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180409